+ All Categories
Home > Documents > A THESIS TITLED - prr.hec.gov.pk

A THESIS TITLED - prr.hec.gov.pk

Date post: 19-Dec-2021
Category:
Upload: others
View: 3 times
Download: 0 times
Share this document with a friend
290
GENE CLONING, CHARACTERIZATION AND THERMODYNAMIC STUDIES OF HIGHLY THERMOSTABLE CELLULOLYTIC ENZYMES FROM GENUS THERMOTOGA A THESIS TITLED NAME: FATIMA AKRAM Registration No. 2013–PhD–BIOT–12 INSTITUTE OF INDUSTRIAL BIOTECHNOLOGYGC UNIVERSITY LAHORE
Transcript
Page 1: A THESIS TITLED - prr.hec.gov.pk

GENE CLONING, CHARACTERIZATION AND

THERMODYNAMIC STUDIES OF HIGHLY THERMOSTABLE

CELLULOLYTIC ENZYMES FROM GENUS THERMOTOGA

A THESIS TITLED

NAME: FATIMA AKRAM

Registration No.

2013–PhD–BIOT–12

INSTITUTE OF INDUSTRIAL BIOTECHNOLOGYGC

UNIVERSITY LAHORE

Page 2: A THESIS TITLED - prr.hec.gov.pk

Gene Cloning, Characterization and Thermodynamic

Studies of Highly Thermostable Cellulolytic Enzymes

from Genus Thermotoga

A THESIS TITLED

Submitted to GC University Lahore

in partial fulfillment of the requirements

for the award of degree of

Doctor of Philosophy

In

Biotechnology

By

FATIMA AKRAM

Registration No.

2013–PhD–BIOT–12

INSTITUTE OF INDUSTRIAL BIOTECHNOLOGY

GC UNIVERSITY LAHORE

Page 3: A THESIS TITLED - prr.hec.gov.pk

IN THE NAME OF

WHO HAS CREATED US

AND

MADE FOR US THE FACULTIES OF

HEARING, SEEING, FEELING

AND

UNDERSTANDING

ALLAH

Page 4: A THESIS TITLED - prr.hec.gov.pk

Declaration

i

DECLARATION

I, Ms. Fatima Akram Registration No. 2013-PhD-BIOT-12 hereby declare that the matter

printed in the thesis titled Gene Cloning, Characterization and Thermodynamic

Studies of Highly Thermostable Cellulolytic Enzymes from Genus Thermotoga is my

own work and has not been submitted and shall not be submitted in future as research

work, thesis for the award of similar degree in any University, Research Institution etc in

Pakistan or abroad.

At any time, if my statement is found to be incorrect, even after my Graduation, the

University has the right to withdraw my PhD Degree.

Dated: 02-06-2017

Signature of Deponent

Page 5: A THESIS TITLED - prr.hec.gov.pk

Plagiarism Undertaking

ii

PLAGIARISM UNDERTAKING

I, Ms. Fatima Akram Registration No. 2013-PhD-BIOT-12 solemnly declare that the

research work presented in the thesis titled “Gene Cloning, Characterization and

Thermodynamic Studies of Highly Thermostable Cellulolytic Enzymes from Genus

Thermotoga”is solely my research work, with no significant contribution from any other

person. Small contribution/help wherever taken has been acknowledged and that

complete thesis has been written by me.

I understand the zero tolerance policy of HEC and Government College University

Lahore, towards plagiarism. Therefore I as an author of the above titled thesis declare that

no portion of my thesis has been plagiarized and any material used as reference has been

properly referred/cited.

I understand that if I am found guilty of any formal plagiarism in the above titled thesis,

even after the award of PhD Degree, the University reserves the right to withdraw my

PhD Degree and that HEC/University has the right to publish my name on

HEC/University website, in the list of culprits of plagiarism.

Dated: 02.06.2017

Signatures of Deponent

Page 6: A THESIS TITLED - prr.hec.gov.pk

Research Completion Certificate

iii

RESEARCH COMPLETION CERTIFICATE

Certified that the research work contained in this thesis titled “Gene Cloning,

Characterization and Thermodynamic Studies of Highly Thermostable Cellulolytic

Enzymes from Genus Thermotoga”has been carried out and completed by Ms.Fatima

Akram Registration No. 2013-PhD-BIOT-12 under my supervision. This Thesis has

been submitted in partial fulfillment of the requirements for the award of Degree of

Doctor of Philosophy in Biotechnology.

Page 7: A THESIS TITLED - prr.hec.gov.pk

Certificate of Approval

iv

Page 8: A THESIS TITLED - prr.hec.gov.pk

Dedication

v

DEDICATED TO

My Supervisor

DR. IKRAM-UL-HAQ (SI), FPAS,

Professor Emeritus, ISESCO Laureate and

Distinguished National Professor,

for his support, encouragement

and help throughout my research work.

Page 9: A THESIS TITLED - prr.hec.gov.pk

Acknowledgements

vi

ACKNOWLEDGEMENTS

Myriads of thanks to Almighty ALLAH, the plenipotent who endowed upon me

potential, courage, resoluteness and enabled me to accomplish this research work, I invoke

peace for Holy Prophet Hazrat Muhammad (Sallaho Alahe Wasallam), who is

forever a torch of guidance for humanity as a whole and showing us the path of knowledge.

There are no words to express sincerest gratitude to my great supervisor Prof. Dr.

Ikram-ul-Haq (Sitara-e-Imtiaz), FPAS, Distinguished National Professor,

Emeritus Professor Institute of industrial Biotechnology, GC University Lahore and

principle investigator of the project entitled “Production of bioenergy from plant

biomass” funded by Ministry of Science & Technology (MoST), Government of Pakistan.

Present research work is the part of this project. I would like to pay my best regards and

deepest gratitude to my impressive supervisor for grant me the opportunity to work in his

project under his dynamic supervision, consolatory behavior and fatherly affection during

the course of research work. His untiring attitude towards work has given me enthusiasm

to follow this approach towards life. I am really fortunate to be a part of his research group.

I could not have imagined having a better supervisor and mentor for my Ph.D. I am highly

thankful to ORIC, GC University Lahore to allot me funds for the completion of my

research work.

I wish to express my gratitude to respected Prof. Naeem Rashid, Director of School of

Biological Sciences (SBS), University of the Punjab Lahore, for his constructive advice,

directions, and guidance during purification of cloned enzymes. I wish to express my

gratitude to respected Prof. Dr. Hamid Mukhtar, Director of Institute of industrial

Biotechnology, GC University Lahore, for his constructive advice, support and professional

guidance. I express my humble gratitude to all my teachers. I would like to thank Mr. Ali

Nawaz, lecturer IIB and express my special thanks to all the authors of the references. I

would also like to thank Ms. Sumra, Ms. Sana, Ms. Saira, Ms. Madeeha Ahmad and Ms.

Iqra (research officers), my research student Muhammad Kaleem deserve special thanks as

Page 10: A THESIS TITLED - prr.hec.gov.pk

Acknowledgements

vii

he was used to stay and work for me till late while I was optimizing the culture conditions.

I would also like to express warm thanks to all the clerical staff of ORIC and IIB specially

Mr. Abdul Sitar and Mr. Habib, and all lab attendants of IIB.

Words are inadequate to express my warmest gratitude to my grandfather, parents,

whose support, motivation and encourage me at each step of life and whose prayers made

it possible for me to carry out my work progressively. My special thanks to my brother Mr.

Wajid Ali, sweet sister and loving cousins for their moral support, always being available

to give positive suggestions and help. Special thanks to all my friends and well-wishers.

Fatima Akram.

Page 11: A THESIS TITLED - prr.hec.gov.pk

Table of Contents

viii

TABLE OF CONTENTS

Declaration………………………………….…………………………………….. i

Plagiarism Undertaking……………………………….………………………...... ii

Research Completion Certificate………….……………………………………... iii

Certificate of Approval…………………….……………………………………... iv

Dedication………………………………….……………………………………... v

Acknowledgements………………………………….……………………………. vi

Table of Contents………………………………….……………………………… viii

List of Tables………………………………….………………………………….. x

List of Illustrations…………………………….………………………………….. xii

List of Abbreviations……………………….…………………………………….. xvii

Abstract………………………………….……………………………………….. xx

CHAPTER NO. 01: INTRODUCTION

1.1. Introduction……….……………………….………………………………….. 01

1.2. Objective……………………………………………………………………… 19

CHAPTER NO. 02: REVIEW OF LITERATURE

2. Review of Literature………………………….………………………………. 20

CHAPTER NO. 03: MATERIAL AND METHODS

3.1. Materials……………………….……………………………………………… 45

3.2. Cloning and Sequencing of TnbglA and TnbglB Genes……………………… 54

3.3. Sub-cloning of TnbglA and TnbglB in Expression Vector…………………… 67

3.4. Heterologous Expression and Production of TnBglA and TnBglB…………… 71

3.5. Purification of Recombinant TnBglA and TnBglB…………………………… 81

3.6. Characterization of Recombinant TnBglA and TnBglB………………………. 85

CHAPTER NO. 04: RESULTS

4.1. β-1,4-glucosidase genes (bglA and bglB) from T. naphthophila …………….. 93

Page 12: A THESIS TITLED - prr.hec.gov.pk

Table of Contents

ix

4.2. Gene Sequence of TnbglA…………………………………………………….. 94

4.3. Gene Sequence of TnbglB…………………………………………………….. 95

4.4. Cloning and Sequencing of TnbglA and TnbglB Genes……………………… 97

4.5. Sub-cloning of TnbglA and TnbglB in Expression Vector…………………… 127

4.6. Expression Analysis of Recombinant Proteins……………………………….. 131

4.7. Preliminary Characterization of TnBglA and TnBglB………………………… 132

4.8. Production of Recombinant TnBglA and TnBglB…………………………….. 137

4.9. Purification of Recombinant TnBglA and TnBglB……………………………. 148

4.10. Characterization of Purified TnBglA and TnBglB…………..……………….. 153

4.11. Structure Analysis of TnBglA and TnBglB………………………………….. 174

4.12. Enhanced Production of Recombinant TnBglA and TnBglB………………... 190

CHAPTER NO. 05: DISCUSSION

5. Discussion………………………………….……………………………….. 204

CHAPTER NO. 06: CONCLUSION

6. Conclusion………………………………….……………………………….. 232

CHAPTER NO. 07: REFERENCES

7. References………………………………….……………………………….. 233

ANNEX

Research Publication and Patent Application...….…………………………. 258

Page 13: A THESIS TITLED - prr.hec.gov.pk

List of Tables

x

LIST OF TABLES

Table Titles Pages

3.1 Composition of amplification reaction mixture. 59

3.2 Compositions of different media. 73

3.3 SDS-PAGE resolving gel composition. 76

3.4 SDS-PAGE Stacking gel composition. 76

3.5 Native resolving/separating gel composition 78

3.6 Native stacking gel composition 79

4.1 Characteristics details of TnbglA and TnbglB genes of T. naphthophila. 94

4.2 (a) List of Non-cutter enzymes for TnbglA.

(b): List of Non-cutter enzymes for TnbglB

99

100

4.3 Oligonucleotides for PCR amplification of TnbglA and TnbglB genes. 101

4.4 (a) Nucleotide sequence BLAST result of TnbglA.

(b) Nucleotide sequence BLAST result of TnbglB.

111

115

4.5 (a) Output data of SignalP 4.1 for TnBglA Signal peptide prediction.

(b): Output data of SignalP 4.1 for TnBglB Signal peptide prediction.

121

122

4.6 Theoretical Properties of TnBglA and TnBglB Protein using ProtParam

tool.

123

4.7 (a) Purification summary of recombinant TnBglA

(b) Purification summary of recombinant TnBglB

150

151

4.8 Substrate specificity of the recombinant TnBglA and TnBglB 166

4.9 (a) Kinetic parameters of TnBglA

(b) Kinetic parameters of TnBglB

168

169

4.10 (a) Kinetic parameters of TnBglA for the hydrolysis of pNPG substrate.

(b) Kinetic parameters of TnBglB for the hydrolysis of pNPG substrate

169

169

4.11 (a) Thermodynamic parameters of TnBglA for pNPG hydrolysis.

(b) Thermodynamic parameters of TnBglB for pNPG hydrolysis.

171

172

4.12 (a) Thermodynamic parameters for denaturation of recombinant TnBglA

(b) Thermodynamic parameters for denaturation of recombinant TnBglB

175

175

Page 14: A THESIS TITLED - prr.hec.gov.pk

List of Tables

xi

4.13 (a) Statistics of Ramachandran Plot. Results computed after Procheck

analysis of TnBglA protein structure using PDBsum

(b) Statistics of Ramachandran Plot. Results computed after Procheck

analysis of TnBglB protein structure using PDBsum

178

180

4.14 (a) Comparison of TnBglA production, induced with 0.5 mM IPTG

(b) Comparison of TnBglA production, induced with 150 mM lactose

201

201

4.15 (a) Comparison of TnBglB production, induced with 0.5 mM IPTG

(b) Comparison of TnBglB production, induced with 150 mM lactose

202

202

Page 15: A THESIS TITLED - prr.hec.gov.pk

List of Illustrations

xii

LIST OF ILLUSTRATIONS

Figures Title Page

1.1 Structure of lignocellulosic plant biomass 04

1.2 Hierarchical structure of cellulose extracted from plants 04

1.3 Schematic representation of biodegradation of cellulosic substrate by using

cellulases

07

1.4 The catalytic retaining two steps mechanism of β-glucosidase for the hydrolysis

of β-glycosidic bond

13

1.5 (a) General structure of Thermotoga

(b) Octopus Spring, water from the main spring supports the growth of a variety

of Thermotoga species

17

1.6 Electron micrographs of stained ultrathin section of Thermotoga naphthophila

RKU-10T strain. Arrows indicate the cell wall (cw) and toga (t)

17

3.1 GeneRuler DNA Ladder Mix, ready-to-use #SM0333 (0.1µg µL-1) 58

3.2 (a) PCR cycling conditions for bglA

(b) PCR cycling conditions for bglB

60

3.3 Restriction map of pTZ57R/T 62

3.4 Map of pET-21a(+) expression vector with cloning and expression sites 68

3.5 Standard curve of Bovine Serum Albumin (BSA) 75

3.6 Novagen Perfect Protein Marker (10-225 kDa), Cat # 69079-3 77

3.7 Standard curve of para-nitrophenol (pNP) 81

3.8 Standard curve of Glucose 89

3.9 Standard curve of Xylose 90

4.1 (a) Linear sequence of TnbglA gene with NEB single cutter endonucleases

(b) Linear sequence of TnbglB gene with NEB single cutter endonucleases

98

99

4.2 Agarose gel of isolated genomic DNA from Thermotoga naphthophila 102

4.3 (a) Analysis of TnbglA amplified product resolved on 0.8% agarose gel

(b) Analysis of TnbglB amplified product after agarose gel electrophoresis

103

103

4.4 Analysis of TnbglA and TnbglB purified PCR product 104

Page 16: A THESIS TITLED - prr.hec.gov.pk

List of Illustrations

xiii

4.5 Blue-white sereening 105

4.6 Randamly selected well-isolated white colonies from master plates 105

4.7 (a) Screening of positive transformants of E. coli DH5α harboring pTZ57R/T–

TnbglA plasmids by colony-PCR

(b) Screening of positive transformants of E. coli DH5α harboring pTZ57R/T–

TnbglB by colony-PCR

106

106

4.8 Isolated recombinant plasmids pTZ57R/T–TnbglA and pTZ57R/T–TnbglB 107

4.9 (a) Restrition analysis of pTZ57R/T–TnbglA plasmid using single restriction

enzyme

(b) Restrition analysis of pTZ57R/T–TnbglB plasmid using single restriction

enzyme

108

108

4.10 (a) Sequence alignment between cloned TnbglA and NCBI retrieved sequence

(b) Sequence alignment between cloned TnbglB and NCBI retrieved sequence

111

114

4.11 (a) Multi-alignment of β-glucosidase from T. naphthophila (TnBglA) with some

other GH1 family β-glucosidases

(b) Multi-alignment of β-glucosidase from T. naphthophila (TnBglB) with some

other GH3 family β-glucosidases

117

120

4.12 (a) Signal peptide prediction graph of TnBglA

(b) Signal peptide prediction graph of TnBglB

121

122

4.13 (a) Analysis of recombinant pTZ57R/T–TnbglA plasmid after double digestion

using two restriction endonucleases

(b) Analysis of recombinant pTZ57R/T–TnbglB plasmid after double digestion

using two restriction endonucleases

124

125

4.14 Streak plate of E. coli DH5α harboring pET-21a(+) 126

4.15 (a) Isolation of plasmid pET-21a(+)

(b) Double restricted pET-21a(+) using restriction endonucleases

126

126

4.16 (a) Purified double restricted pET-21a(+)

(b) Purified double digested DNA fragments (TnbglA and TnbglB)

127

127

4.17 Several white bacterial colonies were appeared on LB-agar plates supplement

with ampicillin after transformation of E. coli DH5α with constructed

expression plasmids

128

Page 17: A THESIS TITLED - prr.hec.gov.pk

List of Illustrations

xiv

4.18 Colony PCR for the screening of positive transformants of E. coli DH5α

harboring constructed recombinant plasmids

128

4.19 Isolated recombinant plasmids treated with RNase 129

4.20 Analysis of recombinant constructed expression plasmids after double digestion

using restriction endonucleases

130

4.21 Several white bacterial colonies were appeared on LB-agar plates supplement

with ampicillin and chloramphenicol after transformation of E. coli BL21

CodonPlus (DE3)-RIPL with recombinant circular plasmids

130

4.22 Colony PCR for the screening of positive transformants of E. coli BL21

CodonPlus harboring constructed recombinant plasmids

131

4.23 SDS-PAGE analysis of recombinant proteins expression in E. coli BL 21

CodonPlus DE3-(RIPL)

132

4.24 Effect of pH and various buffer system on the activity of TnBglA 134

4.25 Effect of temperature on TnBglA and TnBglB 135

4.26 Effect of time course profile of recombinant TnBglA and TnBglB 135

4.27 Cell fractionation analysis for cloned TnBglA and TnBglB 136

4.28 SDS-PAGE analysis of cell lysate (intracellular) and supernatant (extracellular)

fractions of recombinant proteins expressed in E. coli BL 21

137

4.29 Effect of Pre-induction optimal cell density of cloned bacterial on recombinant

enzymes expression

138

4.30 Effect of heat shock treatment on recombinant TnBglA and TnBglB expression 139

4.31 Analysis of different concentration of IPTG as inducer on TnBglA and TnBglB

expression

140

4.32 Effect of different concentration of IPTG on recombinant enzymes expression 141

4.33 Effect of different induction temperature on recombinant TnBglA and TnBglB

expression

142

4.34 Effect of agitation speed on culture growth and recombinant enzymes

expression

143

4.35 Effect of medium pH on the recombinant protein expression 145

4.36 Analysis of TnBglA and TnBglB expression after induction 146

4.37 Effect of incubation time on recombinant enzymes expression 147

Page 18: A THESIS TITLED - prr.hec.gov.pk

List of Illustrations

xv

4.38 (a) SDS-PAGE analysis of TnBglA from pET-21a–bglA in E. coli BL21

CodonPlus (DE3)-RIPL

(b) SDS-PAGE analysis of TnBglB from pET-21a–bglB in E. coli BL21

CodonPlus (DE3)-RIPL

150

151

4.39 Native PAGE analysis of TnBglA and TnBglB 152

4.40 Effect of pH on the activity of purified TnBglA and TnBglB 154

4.41 Effect of temperature on the activity of purified TnBglA and TnBglB 155

4.42 Effect of time course profile of recombinant TnBglA and TnBglB 156

4.43 Thermal stability of TnBglA and TnBglB 157

4.44 pH stability of TnBglA and TnBglB 158

4.45 Effect of metal ions and different inhibitors on purified TnBglA and TnBglB

activity

160

4.46 Effect of organic solvent on purified TnBglA and TnBglB activity 162

4.47 Effect of short chain alcohol on purified TnBglA and TnBglB activity 163

4.48 Effect of glucose and xylose on purified TnBglA and TnBglB activity 165

4.49 (a) Lineweaver-Burk plot for the hydrolysis of various substrates using TnBglA

(b) Lineweaver-Burk plot for the hydrolysis of various substrates using

TnBglB.

167

168

4.50 Arrhenius plot of recombinant enzymes of T. naphthophila 171

4.51 Pseudo-first-order plots for irreversible thermal denaturation of recombinant

thermostable enzymes

173

4.52 Arrhenius plot for irreversible thermal inactivation of purified recombinant

enzymes

174

4.53 (a) Secondary structure of TnBglA

(b) Secondary structure of TnBglB

176

177

4.54 (a) Ramachandran Plot for TnBglA on Procheck using PDBsum

(b) Ramachandran Plot for TnBglB on Procheck using PDBsum

178

179

4.55 (a) Three-dimensional (3-D) model structure of TnBglA

(b) Catalytic triad of TnBglA

(c) Three-dimensional (3-D) model structure of TnBglB

(d) Catalytic triad of TnBglB

181

182

183

184

Page 19: A THESIS TITLED - prr.hec.gov.pk

List of Illustrations

xvi

4.56 Ligplot output for H-bond interactions of amino acid residues of TnBglA

(a) Involved in binding with cellobiose

(b) With the p-phenyl sugars as substrates

(c) Surface view of 3-D structure of TnBglA

185

186

187

4.57 Ligplot output for H-bond interactions of amino acid residues of TnBglB

(a) Involved in binding with cellobiose

(b) With the p-phenyl sugars as substrates

(c) Surface view of 3-D structure of TnBglB

188

189

189

4.58 Effect of media on recombinant enzymes activity induced with 0.5 mM IPTG 191

4.59 Effect of media on recombinant enzymes activity induced with 150 mM lactose 192

4.60 SDS-PAGE analysis of recombinant TnBglA in E. coli BL 21 grown in various

0.5 mM IPTG inducing media

193

4.61 SDS-PAGE analysis of TnBglB in E. coli grown in various IPTG inducing

media

194

4.62 SDS-PAGE analysis of TnBglA in E. coli grown in various lactose inducing

media

195

4.63 SDS-PAGE analysis of TnBglB in E. coli grown in various lactose inducing

media

196

4.64 Effect of IPTG on recombinant enzymes expression in 4×ZB medium 198

4.65 Effect of lactose on the expression of recombinant TnBglA and TnBglB

enzymes in 4×ZB medium

199

4.66 SDS-PAGE analysis of recombinant purified TnBglA and TnBglB after lactose

(150 mM) induction in 4×ZB medium

203

Page 20: A THESIS TITLED - prr.hec.gov.pk

List of Abbreviations

xvii

LIST OF ABBREVIATIONS

AEC Anion exchange chromatography

Ag Silver

ANOVA Analysis of variance

Aͦ Angstrom

APS Ammonium per sulphate

Asp/D Aspartic acid

α Alpha

Ba Barium

β Beta

BGL/Bgl Beta-glucosidase/β-glucosidase

BLAST Basic Local Alignment Search Tool

BME Beta-mercaptorthanol

Bp Base pair

BSA Bovine serum Albumin

Ca Calcium

CaCl2 Calcium chloride

Cal Calorie

°C Degrees Celsius

CAPS 3-(cyclohexylamino)-1-propanasulfonic acid

CH3CO2K Potassium acetate

CMC Carboxymethyl cellulose

Conc. Concentration

C-Score Raw cleavage site score (for signal peptide determination)

Cu Copper

Da Daltons

ddH2O Double distilled water

dH2O Distilled water

DCW Dry cell weight

DNA Deoxyribonucleic acid

DNS 3,5-Dinitrosalicylic acid

dNTPs Deoxyribonucleoside triphosphates

D-Score Discrimination score

DTT Dithiothreitol

EDTA Ethylene diamine tetraacetic acid

ExPASY Expert Protein Analysis System

Fe Iron

G Gibb’s free energy

g/L Grams per liter

GHs Glycosyl hydrolase(s)

Page 21: A THESIS TITLED - prr.hec.gov.pk

List of Abbreviations

xviii

Glu/E Glutamic acid

H Enthalpy

h Hours(s)

HEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

HIC Hydrophobic Interaction chromatography

His/H Histidine

IEC/IEXC Ion exchange chromatography

IPTG Isopropyl-β-D-thiogalactopyranoside

J Joule

K Kelvin

kcat Turnover number

kcat/Km Catalytic efficiency

kb Kilo base pair/1,000 bp

Kd Dissociation constant

kDa Kilo Daltons

Ki Inhibitor constant

Km Michaelis-Menten constant

L Liter

LB Luria-Bertani

lb/in2 Pounds per square inch (psi)

Max/max Maximum

MCS Multiple cloning sites

MES 2-(N-morpholino)-ethanesulfonic acid

Mg Magnesium

mg/mL Milligram per milliliter

MgCl2 Magnesium chloride

MgSO4 Magnesium Sulphate

Millivolt(s) mV

min Minute(s)

mL Milliliter

mM Millimolar

µg Microgram

µL Microliter

mol Mole

MOP 3-(N-morpholino)-propanesulfonic acid

MW Molecular weight

MWCO Molecular weight cutoff

NaCl Sodium chloride

NCBI National Center for Biotechnology Information

ng Nanogram

Ni Nickel

Ni-column Nickel column

Page 22: A THESIS TITLED - prr.hec.gov.pk

List of Abbreviations

xix

nm Nanometer

OD Optical density

oNPG o-nitrophenyl-β-D-glucopyranoside

oNPGal o-nitrophenyl-β-D-galactopyranoside

ORF Open reading frame

Pb Lead

% Percentage

PCR Polymerase Chain Reaction

pH potential of Hydrogen/ power of Hydrogen

pI Isoelectric point

pM picomole

PMSF Phenylmethylsulfonyl fluoride

pNPF p-nitrophenyl-β-D-fucopyranoside

pNPG/p-β-NPG p-nitrophenyl-β-D-glucopyranoside

pNPGal p-nitrophenyl-β-D-galactopyranoside

pNPMan p-nitrophenyl-β-D- mannopyranoside

pNPX p-nitrophenyl-β-D-xylopyranoside

Rb Rubidium

RPM/rpm Revolution per minute

S Entropy

SDS-PAGE Sodium dodecyl sulfate polyacrylamide gel electrophoresis

sec Second (s)

SPSS Statistical package for the social sciences

S-Score Signal peptide score (for signal peptide determination)

TAE Tris acetic acid EDTA

Taq Thermus aquaticus

TCP Total Cell Protein

TE Buffer Tris EDTA Buffer

TEMED N,N,N',N'-Tetra methyl ethylene diamine

Tm Melting temperature

U/mL Units per milliliter

U/mL/min Units per milliliter per minute

Vmax Maximal velocity

w/v Weight per volume

X-gal 5-bromo-4-chloro-3-indolyl-β-D-galactopyranoside

Y-Score Combined cleavage site

Zn Zinc

Page 23: A THESIS TITLED - prr.hec.gov.pk

ABSTRACT

Page 24: A THESIS TITLED - prr.hec.gov.pk

Abstract

xx

Abstract

With a paradigm shift in industry, moving from natural fuels to alternative renewable

resource utilization, concisely the growing demands of bioenergy has led to the emphasis

on novel cellulolytic enzymes to improve efficiency of bioconversion process of

lignocellulosic plant biomass. Currently, biodegradation is an area of extensive research,

the cynosure of biofuel industry is on the utilization of non-edible lignocellulosic biomass

(feedstock, agricultural and municipal residues) as an exploitable, inexpensive and

potential source of alternative renewable energy in the form of bioethanol, thus the need of

efficient thermostable cellulases are expected to increase in the future. β-glucosidase is an

essential member of cellulase enzyme system that plays a critical role in cellulosic substrate

hydrolysis and in many other biological processes.

Therefore, the present study describes cloning of two novel highly thermostable

cellulolytic enzymes β-1,4-glucosidases (TnBglA and TnBglB) from a bacterium

Thermotoga naphthophila RKU-10T and overexpressed in Escherichia coli BL21

CodonPlus (DE3)-RIPL. Purification and biochemical characterizations together with

kinetic and thermodynamic analysis give insights about the thermostability of both

enzymes. Various cultivation and induction strategies were applied to enhance the

production of engineered host cells density and expression of highly efficient

thermotolerant TnBglA and TnBglB, induced individually with IPTG and an alternative

inducer lactose. Culture conditions and other parameters including media compositions,

pre-induction optical density, agitation, inducer concentrations, temperature and time of

induction were optimized to achieve maximum yield of heterologous proteins.

Genomic DNA of T. naphthophila was used as template to amplify two cellulolytic genes

TnbglA and TnbglB (ADA66698.1 and ADA66752.1) of 1.341 and 2.166 kb, which

encoded β-1,4-glucosidase proteins of 446 and 721 amino acid residues, respectively.

Amplicons of genes were cloned initially in pTZ57R/T vector by employing dA×dT tailing

technique and consequently subjected to sequence analysis. Sequence homology analysis

demonstrated that TnBglA and TnBglB belong to glycoside hydrolase family 1 (GH1) and

family 3 (GH3), respectively. Further, both genes were sub-cloned in pET-21a(+) vector

Page 25: A THESIS TITLED - prr.hec.gov.pk

Abstract

xxi

and over-expressed in E. coli BL21 under the control of inducible T7 lac promoter.

Initially, LB medium was used for the production and optimization of various cultivation

parameters to get the high level expression of both recombinant enzymes. However,

optimal expression and activity of both enzymes were observed when culture induced with

0.5 mM IPTG after heat shock treatment (42°C, 1 h) at 0.6 pre-induction optical density

(OD600nm) followed by incubation at 22°C for 72 h in a shaking incubator at 200 rpm.

Both extracellular TnBglA and TnBglB with a molecular weight of 51.50 and 81.14 kDa,

respectively were purified to homogeneity by ion-exchange and hydrophobic interaction

chromatography after heat treatment at 70°C for 1 h. Purified enzymes TnBglA and TnBglB

displayed optimal activity at pH 7.0 (95°C) and pH 5.0 (85°C temperature), respectively.

Both enzymes were quite stable over a broad range of pH (6.0-8.5) and temperature (60-

90°C), fairly stable up to 8 h at 80°C. GH1 TnBglA activity was stimulated in the presence

of glucose and xylose, with a Ki value of 1200 and 1300 mM, respectively. Whereas, GH3

TnBglB revealed Ki value of 150 mM for glucose and 200 mM for xylose, both enzymes

displayed affinity towards p-nitrophenyl substrates and cellobiose.

Km, Vmax and kcat values of TnBglA, using pNPG as substrate, were 1.5 mM, 297 mmol mg-

1min-1, and 1527778 s-1, respectively. Whereas, TnBglB showed Km, Vmax and kcat values of

0.45 mM, 153 mmol mg-1min-1 and kcat 1214285.7 s-1, respectively using pNPG as

substrate. Thermodynamic parameters as ∆H*, ∆G* and ∆S* for pNPG hydrolysis by

TnBglA were calculated at 95°C as 25.7 kJ mol-1, 47.24 kJ mol-1 and -58.6 J mol-1 K-1,

respectively. Thermodynamic parameters for pNPG hydrolysis by TnBglB like ∆H*, ∆G*

and ∆S* were calculated at 85°C as 24.1 kJ mol-1, 46.55 kJ mol-1 and -62.74 J mol-1 K-1,

respectively. TnBglA displayed a half-life (t1/2) of 5.21 min at 97°C with denaturation

parameters of enzyme including ΔH*D, ΔG*D and ΔS*D were 662.04 kJ mol-1, 110.10 kJ

mol-1 and 1.491 kJ mol-1 K-1, respectively. TnBglB showed a half-life (t1/2) of 4.44 min at

94°C with denaturation parameters of enzyme including ΔH*D, ΔG*D and ΔS*D were

283.78 kJ mol-1, 108.7 kJ mol-1 and 0.477 kJ mol-1 K-1, respectively.

Generally, inadvertently preparing medium and unintentional induction of engineered E.

coli BL21, give poor or variable yields of heterologous proteins. Therefore, to enhance the

Page 26: A THESIS TITLED - prr.hec.gov.pk

Abstract

xxii

activity and production of an industrially relevant cloned enzymes through various

cultivation and induction strategies. High-cell-density and optimal heterologous proteins

expression were obtained in 4×ZB medium after 72 h inducement at 22°C when culture

gave heat shock (at 42°C for 1 h) at 0.6 OD600nm and induced either with 0.5 mM IPTG/150

mM lactose. TnBglA and TnBglB activities were enhanced 3.8 and 0.096 fold in 4×ZB

medium with 150 mM lactose, respectively under optimal conditions. However,

considerably greater dry cell weight of TnBglA and TnBglB cultures were 11.30 g DCW

L-1 and 11.08 g DCW L-1 in 4×ZB medium, respectively induced with 150 mM lactose.

Use of the inexpensive and non-toxic inducer lactose, and an effective process strategy is

essential to achieve a high level of enzyme expression.

The expression and purification scheme, presented here, has a potential of scaling up to

obtain pure and active enzymes, relatively economical for further studies and other

applications. Finally, this is a report on enhanced production of highly heat active cloned

β-glucosidases from T. naphthophila (TnBglA and TnBglB) with high catalytic efficiency

and low product inhibition, which have excellent tolerance against glucose and xylose, and

also exhibit independence of detergents, chemical inhibitors and metal cations. All these

significant features make both TnBglA and TnBglB appropriate candidates for

biotechnological and industrial applications.

Keywords: thermostable; β-glucosidase; thermodynamics; glucose tolerance;

heterologous protein expression; induction strategy; Thermotoga naphthophila.

Page 27: A THESIS TITLED - prr.hec.gov.pk

CHAPTER-I

INTRODUCTION

Page 28: A THESIS TITLED - prr.hec.gov.pk

Introduction

1

1.1. Introduction

The widening gap between energy demand and supply has led to power outages, and has

caused disruption in the economy and development of countries. Despite having abundant

renewable energy resources, the sharp growing demand has led to countrywide energy

crisis which threatens the economic growth. Overall energy consumption, impending

shortages of natural fossil fuels and global warming are expected to increase in near future.

Every year world energy demand is rapidly increasing, and in 2012 energy consumption

has increased by 1% and by this carbon dioxide (CO2) emissions has led to an augmented

by 1.4% (Enerdata, 2013). The petroleum-based fossil fuels are the main resource of energy

in this world for the industrialization and economic growth of states. Globally, all known

and anticipated reserves of oil will not significantly run out for more than 100 years because

of exceed production and high consumption rates (Saratale and Oh, 2012).

The use of fossil fuels have various multi-faceted problems like supply instability, rising

cost, and global warming by a steady rise in environmental pollution which may adversely

affect Earth planet in near future. These economic and geopolitical factors have played a

significant role in reviving an interest in renewable energy resources that can decrease and

displace our reliance on uncertain and unstable sources of petroleum-based liquid fuels for

world-wide major energy demand, and also decline change in global climate (Kuhad et al.,

2016). Therefore, the search for more sustainable energy sources which will be ecofriendly,

inexpensive and high in yield, is now no longer something of a luxurious (Khelil and

Cheba, 2014). These concerns have shifted researcher’s efforts to develop technologies for

the production of ‘greener energy’ known as bioenergy that is an alternative, sustainable,

clean and renewable energy source, which can effectively use in place of fossil fuel to meet

rising energy demands of the world (Maki et al., 2009; Raghuwanshi et al., 2014).

Biofuels (bioenergy) are the only substitute source of energy for the predictable future and

can form the basis of sustainable development in terms of environmental and

socioeconomic concerns (Demirbas, 2007). Biofuels are solid, liquid (bioethanol,

biobutanol and biodiesel) or gaseous (hydrogen and methane) fuels acquired relatively

from recently dead biological material mainly from plant biomass (agricultural byproducts

Page 29: A THESIS TITLED - prr.hec.gov.pk

Introduction

2

and dedicated energy crops) while fossil fuels are derived from long dead biological

material (Mohanram et al., 2013). Firstly, the idea of converting natural plant biomass-

derived carbohydrate sugars to biofuel was proposed in the 1970s (Lamers et al., 2008). In

recent time, this idea is being considered again seriously and biofuel has received

considerable attention because it offer a means to minimize dependence on natural crude

oil and to reduce emissions of greenhouse gases (CO, CO2 and CH4) which affect our

environment inadequately. Policy intercessions, in the form of supports and mandated

biofuels blending with fossil fuels are driving the rush to liquid biofuels.

Biofuel can contribute to develop an economic, and to make the environment better as it

contains higher octane ratings ethanol which combust in a more efficient and clean way

than gasoline, consequently their atmospheric carbon footprint is integrally decreased as it

offers carbon neutral alternative (Mohanram et al., 2013). Many developed as well as

developing countries (states) have made huge investments in infrastructure, process

development and biofuel (bioethanol) production facilities (Koppram et al., 2014). In 2008,

1.8% world transport fuel has provided by biofuel (Bringezu et al., 2009). However,

petroleum consumption can immediately be decreased by blending the fuel with

bioethanol, and the use of blended gasoline can cut down CO2 emissions by 25-30%

(Mohanram et al., 2013). In US, Canada, and Brazil renewable fuel standard has been

boosted, in these days, 10-25% ethanol successfully mixed with fuel (Maki et al., 2009).

According to an international report, Pakistan can instantaneously reduce its petrol

consumption by at least 30-40 million barrels per year, by blending fuel with 10% ethanol.

On the Earth's surface, plants use solar energy to fix atmospheric CO2 and collectively

resolved to recycle an estimated 1011 tons of carbon per year, which is used in the formation

of complex carbohydrates and their derivative via photosynthesis (Yeoman et al., 2010).

For the commercial production of biofuels, several plants and plant-derived materials

(sugars) are used as substrates including grains for ‘first generation’, and lignocellulosic

waste biomass for ‘second generation’ biofuels. Currently in biofuel industries, sucrose-

and starch- containing agricultural crops (sugarcane, corn, maize and wheat) are used as

substrates for production with conventional technologies, known as first generation

biofuels.

Page 30: A THESIS TITLED - prr.hec.gov.pk

Introduction

3

However, edible raw materials are a controversial resource for biodegradation and will not

be adequately sufficient to meet the raising demand of biofuel (Greene et al., 2004;

Mohanram et al., 2013). First generation biofuels production, neither economically nor

ecologically sustainable, as grain substrates require large land areas for cultivation and

compete with crops (edible plant materials) meant for human consumption (Yeoman et al.,

2010). Therefore, there is a robust necessity to look at non-consumable lignocellulosic

plant biomass (feedstock, municipal and agricultural wastes) that can use effectively as

cellulosic substrates for the production of second generation biofuels and commodity

chemicals, this renewable natural carbohydrates will provide us a sustainable and

economical alternative energy in the form of bioethanol (Klose et al., 2012).

‘Second generation’ of biofuel is highly imperative because they are based on highly

abundant and inexpensive lignocellulosic biomass; a complicated composite with three

main biopolymers i.e., approximately 40-55% cellulose, 25-50% hemicellulose (xylan) and

10-40% of lignin, although individual compositions vary among species. (Vanitha et al.,

2011; Mohanram et al., 2013). Non-edible cellulosic biomass is a significant and

noteworthy energy source to cope with the food problem and shortage of energy with

explosive or abrupt increase in human population. Cellulosic wastes are the chief source of

biomass which constitutes switch grasses, miscanthus, forestry and agricultural residues

(sugarcane bagasse, corn fiber, rice hulls and wheat bran etc.), municipal and industrial

superfluous materials. These raw materials are cheap, unexploited, environmentally sound,

easily accessible, and inexhaustible biological resource which could be proficiently utilized

as substrate for bioethanol production instead of edible grain crops (Maki et al., 2009;

Yennamalli et al., 2013; Koppram et al., 2014).

Cellulose is the most abundant (profuse) organic compound on Earth, with the formula

(C6H10O5)n, virtually inexhaustible and a great potential renewable bioenergy source

(Teugjas and Väljamäe, 2013); major constructive component of algae and plant cell walls,

although some bacteria and marine invertebrate animals (tunicates) also produced cellulose

(Väljamäe et al., 2001; Lynd et al., 2002; Tang et al., 2014). Cellulose, a bio-polymer is

mingled with hemicellulose and lignin elements which make a complex architecture of

plant cell wall (Figure 1.1). Cellulose exist in the form of microfibrils, a compact inflexible

Page 31: A THESIS TITLED - prr.hec.gov.pk

Introduction

4

highly organized unbranched homo-polysaccharide composed of D-glucose units linked

via β-1,4-glycosidic bond (Figure 1.2) (Artzi et al., 2014). Many of these polysaccharide

linear chains are bound together through Van der Waals forces and hydrogen bonding,

arranged in a parallel arrays and in turn form macrofibrils. Cellulose is different in their

configuration has crystalline domain which intermixed with less ordered amorphous

regions, where these forces are loosen (Fernandes et al., 2011; Endler et al., 2011).

Figure 1.1: Structure of lignocellulosic plant biomass (Tomme et al., 1995).

Figure 1.2: Hierarchical structure of cellulose extracted from plants (Rojas et al., 2015).

Page 32: A THESIS TITLED - prr.hec.gov.pk

Introduction

5

Cellulose degradation is challenging due to its multifaceted and recalcitrant nature (Dong

et al., 2010). Generally, production of bioethanol involves pretreatment of cellulosic plant

biomass and its enzymatic conversion to fermentable sugars (glucose) molecules which

can be converted to bioethanol (Chan et al., 2016). Bioethanol has been conceived of as

the future source of green alternative for the development of transportation fuels and

various other chemicals (Kuhad et al., 2016). This process has many other application,

such as treating waste water from textile, laundry detergents, dying and pulping industries.

(Wang et al., 2015). The limiting factor of bioethanol production is the efficient conversion

of biomass to fermentable sugars; however, complex and rigid structure of cellulose is a

leading bottleneck to preclude the development of a lucrative production method.

Initially, hydrolysis of cellulosic material was performed chemically at high temperature

using various acids but this approach was often non-specific, slow and some time the

process became uncontrolled which is the major barrier to its widespread application (Kim

et al., 2001). In nature, cellulosic material is hydrolyzed mostly by the hydrolytic and

oxidative biocatalysts of bacteria and fungi (Lynd et al., 2002; Horn et al., 2012). Though,

several conventional (thermal and chemical) and non-conventional (biochemical and

microbial) strategies have been suggested and employed, either alone or in combination

but none have proven appropriate and satisfactory as a standalone strategy because of the

intricacy and complexity of feedstock (biomass). This obstacle needs robust technologies

for bioconversion which are insensitive to fluctuation (variation) in bio-feedstock,

adequately vigorous to face biologically challenging process operating conditions and can

significantly increase the yield (Sohel and Jack, 2012).

Keeping all these specifications in mind, plausibly it is better to consider thermophilic and

hyperthermophilic microorganisms (fungi and bacteria that natively live at extremely high

temperatures) and their biocatalysts for decisive roles in saccharification process

(hydrolysis of a polysaccharides or cellulose into soluble sugar components). Recently a

large number of thermophiles and hyperthermophiles are available having an extensive and

competent glycosidic hydrolase (GH) inventory (VanFossen et al., 2008). Consequently,

biodegradation using highly effective thermo- efficient cellulolytic enzymes is the most

promising conversion technology. Thermostable cellulases require less energy and mild

Page 33: A THESIS TITLED - prr.hec.gov.pk

Introduction

6

environmental conditions to convert biomass into fermentable sugars that are used to make

many important bio-based industrial products, which can replace fossil fuels (Zhang et al.,

2007; Vanitha et al., 2011).

Cellulases are the third largest group of industrial biocatalysts, have been commercially

available for more than 30 years. Applied and basic studies on cellulases have

demonstrated their industrial and biotechnological applications in various fields.

Commonly, cellulases have been extensively used in textile (for biofinishing, biopolishing

of fabrics and biostoning of jeans), food processing, animal feed, fruit juices, brewing (beer

purification) and wine making, to enhance flavor and aroma in brewery products (beer,

wine, fruit juices, tea and coffee), in agriculture to control plant diseases and pests, to

enhance plant growth and seed germination, to enhance oil extraction yields, to improve

the nutritive quality of bakery products, in laundry detergents, in paper and pulp (to

increase the brightness of paper and to decrease the viscosity of pulp), cosmetic and

pharmaceutical industries. Most attractively have developed significant interest due to their

auspicious and promising applications in biofuel production (Kuhad et al., 2011; Ferreira

et al., 2014).

Enzymatic saccharification of cellulosic substrates is a complex and intricate process which

requires the synchronized (coordinated) action of three main cellulolytic enzymes: (i) endo-

1,4-beta -glucanases (EGs, EC 3.2.1.4), (ii) cellobiohydrolases also called exo-1,4-beta-

glucanase (CBHs, EC 3.2.1.91), and (iii) beta-1,4-glucosidases (Bgls, EC 3.2.1.21), these

three hydrolytic enzymes are usually grouped as cellulase system (Colussi et al., 2012;

Singh et al., 2016). Firstly, endo-1,4-β-glucanases (EGs) randomly cleave the linear chains

of cellulosic fibers in amorphous regions (middle portion of cellulose molecules), where

EGs are easily accessible, act on the inner β-1,4-glucosidic bonds and convert cellulosic

polysaccharide chain into short fragments of different lengths. Then, cellobiohydrolases

(CBHs) act at the exposing sites of both reducing and non-reducing ends of these short

chains in less-accessible crystalline regions to release the cellodextrin and cellobiose as

major products, which finally hydrolyze by β-1,4-glucosidases (Bgls) to produce glucose

monomers (Figure 1.3) as the final product of biodegradation (Sørensen et al., 2013;

Page 34: A THESIS TITLED - prr.hec.gov.pk

Introduction

7

Dantur et al, 2015; Pereira et al, 2015; Singh et al., 2016). During hydrolysis, activities of

endo-1,4-β-glucanase and cellobiohydrolase are inhibited by cellobiose (product inhibitor),

therefore, β-glucosidase stimulates and regulate the cellulose hydrolysis more efficiently

by relieving cellobiose inhibition (Liu et al., 2012; Pei et al., 2012; Schroder et al., 2014).

Figure 1.3: Schematic representation of biodegradation of cellulosic substrate by using cellulases. During

hydrolysis cellobiose and cello-oligosaccharides are produced by synergistic action of cellobiohydrolases

(exo-acting enzyme, CBHs) and endo-1,4- β-glucanases (endo-acting enzyme, EGs), which are subsequently

hydrolyzed by β-glucosidases to produce glucose units (monomers) as a final product of complete cellulose

metabolism (Singh et al., 2016).

β-1,4-glucosidases (β-D-glucoside glucohydrolase) are ubiquitous enzymes, widely

distributed in nature, can be found in simple microbes (fungi and bacteria) to highly

evolved plants and animals, and considered to be a significant component of cellulase

system (Krisch et al., 2010). It acts on cellobiose, alkyl- and aryl-β-D-glucosides in β-1,4-

glucosidic linkage, exhibits transglycosylase and alkyl transferase activities, and catalyzes

the synthesis of glycoconjugates and alkyl glucosides (Teugjas and Väljamäe, 2013). β-

glucosidases are broadly used in several fundamental processes ranging from

Page 35: A THESIS TITLED - prr.hec.gov.pk

Introduction

8

developmental regulation (metabolic pathways) to chemical protection against pathogen

(insects and microbes) attack, and a number of biotechnological and industrial applications,

including bioethanol production from agricultural cellulosic wastes, host-pathogen

interaction, oncogenesis, cellular signaling, synthesis of valuable β-glucosides and

production of biodegradable nonionic surfactants (Bhatia et al., 2002; Chuankhayan et al.,

2007; Li et al., 2013).

β-glucosidase has achieved significant attention due to their diverse and essential

physiological roles in various biological system that depend on the enzyme location in

which they present (Singhania et al., 2012). In cellulolytic microorganisms, β-glucosidase

is the main biocatalyst for the metabolism of carbohydrate (grow on various glucose

conjugates or disaccharides and oligosaccharides sugars) and cellulase induction, plays

crucial role in cell wall mechanism, symbiotic association and host-pathogen (Singh et al.,

2016). In plants, β-glucosidase plays pivotal roles in several essential biological (natural)

processes such as in pigment metabolism, cell wall development, fruit ripening, seed

development, detoxification of cyanogenic glycosides, regulating chemical defense against

pathogen attack by releasing pathogen-defending compounds from glycosidic bonds,

production and activation of phytohormones, and exhibits a vital role in hydrolysis of

glycosylated flavonoids (Belancic et al., 2003; Marotti et al., 2007; Krisch et al., 2010;

Mehmood et al., 2014). In mammals, β-glucosidase involves in degradation of glycolipids

in mammalian lysosomes and hydrolysis of glucosyl ceramides, thus deficiency of this

enzyme causes a non-neuropathic lysosomal storage disorder in human known as

Gaucher’s disease (Lieberman et al., 2007; Singhania et al., 2012).

β-glucosidase has large potential applications in various industries, gained more attention

due to liberation of aromatic compounds and flavor from tasteless glycosylated precursors

present in fermenting products, musts, fruits juices and wine (Su et al., 2010; Fan et al.,

2011). β-glucosidases are extensively used in food processing, dairy, beverage and flavor

industries. It is significantly used in wine making, production of fruit juices, and

extensively applied to improve the flavor and aroma of fruit juices, wines and tea (Pei et

al., 2012; Keerti et al., 2014; Cota et al., 2015). During juice extraction, hydrolysis of

naringenin to pruning by enzyme reduced the bitterness of juices and liberate aroma from

Page 36: A THESIS TITLED - prr.hec.gov.pk

Introduction

9

grapes of wine (Harhangi et al., 2002). Aside from the enhancement of flavor and aroma,

β-glucosidases may increase the nutritional value of beverages, food and feed by release

of anti-oxidants, vitamins and other valuable compounds from their glycosides (Singh et

al., 2016).

In tea industry, the essential oil contents has been improved by treating with β-glucosidase

and enhanced the quality of beverages products (Krisch et al., 2010; Su et al., 2010). In

rice, enzymatic action of β-glucosidase can be released vitamin B6 (pyridoxine) from

pyridoxine glucoside (Opassiri et al., 2004). It mostly used to extract the medically

essential compounds from vegetables and fruits by the hydrolysis of phenolic and

phytoestrogen glucosides (Schroder et al., 2014). In cellulosic-based feeds used as an

additives, supplementation of β-glucosidase is highly beneficial for single-stomach animals

like chickens and pigs which enhanced the digestibility of feed and lead to better utilization

of nutrient (Krisch et al., 2010).

Production of aglycone by the breakdown of soybean isoflavone glycosides via enzyme β-

glucosidase is most desired application of this enzyme in industries. Isoflavones are

recognized to prevent several chronic diseases (improve bone health and lowers risks of

cardiovascular diseases) and certain cancers because they potently inhibit the growth of

cancer cells. Isoflavones, in soy-based foods are chiefly present in inactive form of

glycosides, and has been proved that the biological effects occur due to aglycone form

rather than their glycosylated form because aglycone forms quickly absorbs by intestine

(Izumi et al., 2000; Hu et al., 2009). This enzyme can be used in the manufacturing of

cosmetics products for melanogenesis inhibitory activities, arbutin-β-glycosides was

synthesized through the transglycosylation reaction using β-glucosidase to make a new

skin whitening agent (Jun et al., 2008).

β-glucosidase is highly employed for the synthesis of oligosaccharides (short chain sugars)

and alkyl-glycosides; oligosaccharides can be applied as growth promoting agent,

diagnostic tools and therapeutic agents, have imperative functions in various biological

systems such as cell proliferation, fertilization, and embryogenesis. Alkyl-glycosides have

several antimicrobial properties and prospective application in cosmetic, chemical,

Page 37: A THESIS TITLED - prr.hec.gov.pk

Introduction

10

pharmaceutical, detergent and food industries as these hydrolyzed and catalyzed by β-

glucosidase enzyme (Bankova et al., 2006; Singh et al., 2016). Furthermore, this versatile

enzyme can be employed in many synthetic reactions because of its wide and varied roles

in nature (Bhatia et al., 2002).

β-glucosidases are a large group of versatile and flexible hydrolytic biocatalysts. According

to different criteria, these heterogeneous enzymes have been categorized into various

groups because there is no single precise way for their classification. Generally, two well-

known strategies have been reported in the literature for the classification of β-glucosidases

by which they can be divided, on the basis of their substrate specificity and

nucleotides/amino acids sequence identity (Henrissat and Bairoch, 1996). Substrate

specificity classifies β-glucosidases into three main groups: (i) aryl-β-D-glucosidases, have

great affinity to aryl-β-D-glucosides, (ii) cellobiases, preferably hydrolyze only

oligosaccharides, and (iii) broad specific β-glucosidases, can hydrolyze various types of

substrates. Most of the characterized β-glucosidases are assembled in third group due to

their catalytic activity towards various substrates (Pei et al., 2012; Fang et al., 2014; Chan

et al., 2016). The most accepted method of classification is on the basis of amino acid

sequence and structural folding similarity (homology), therefore β-glucosidases have been

sub-divided into various glycoside hydrolases (GHs) families 1, 3, 5, 9, 30 and 116

(Sørensen et al., 2013; Lombard et al., 2014).

However, β-glucosidases are commonly found in either the glycoside hydrolase (GH)

family 1 or family 3 (Opassiri et al., 2007; Cantarel et al., 2009). Currently, 133 GH

families are listed (registered) in the regularly updated website (http://www.cazy.org)

Carbohydrate Active enZyme (CAZy) (Cantarel et al., 2009). Structural features and amino

acid sequence of enzymes are the more informative and important tool for classification,

therefore International Union of Biochemistry and Molecular Biology categorized GH

families into structurally determined groups (Harris et al., 2010). Enzyme’s structural

features can be applied to analyze and study the topographies of other group members of

family by using various bioinformatics tools and approaches. Particularly, tertiary structure

of enzyme at active site dictates their catalytic behavior (mechanism) and affinity towards

various substrates. Enzymes which have similar sequence and conserved motifs, are

Page 38: A THESIS TITLED - prr.hec.gov.pk

Introduction

11

assemble in the same GH family. Additionally, GH families are categorized into various

clans, families with conserved catalytic amino acid residues, analogous catalytic

mechanism and domain structures, suggestive and evocative of a mutual ancestry (lineage),

are gathered in single clan (Singh et al., 2016).

Large number of families have grouped in Clan GH-A including β-glucosidase GH family

1, GH family 5 and GH family 30, they all have a conserved (α/β)8 triose phosphate

isomerase (TIM) barrel scaffold or protein domain that has catalytic site. Maximum

number of characterized β-glucosidases so far contains a classic (α/β)8 barrel structure that

is a key feature of GH family 1 (GH1) and their catalytic active site have two conserved

carboxylic acid (glutamic acid, Glu166 and Glu351) residues on four and seven β-strands,

acting as catalytic acid/base (proton donor) and nucleophile, respectively (Cairns and Esen,

2010; Vuong and Wilson, 2010). The structure of β-glucosidases from Phanerochaete

chrysosporium (a basidiomycete) and human cytosolic have been studied by x-ray

crystallography and finally illustrated the presence of (α/β)8 barrel folds (Nijikken et al.,

2007; Tribolo et al., 2007).

Structural data of GH family 3 (GH3) β-glucosidases are still scarce, since only few have

been characterized thoroughly up till now. β-glucosidases from GH3 have two conserved

domains, an (α/β)8 barrel and α/β sandwich domain including a six stranded β-sheet

sandwich between three α-helices on both/either side (α/β)6. Conserved active site of GH3

β-glucosidases are located at the interface of both sandwich domains, however each domain

contributes one carboxylic acid catalytic amino acid residue, at the N-terminal domain an

aspartic acid (Asp285) and at the C-terminal domain a glutamic acid (Glu491) act as a

catalytic nucleophile and a proton donor, respectively (Singhania et al., 2012; Colussi et

al., 2015).

An alternative β-glucosidases classification system have been established on sequence

homology, and allocated these enzymes into two sub-families: (i) β-glucosidase family A

contains phospho-β-glucosidases and β-glucosidases from microorganisms to mammals,

and (ii) β-glucosidase family B comprises enzyme from rumen bacteria, molds and yeasts.

GH family 1 comprises approximately 62 β-glucosidases from archaebacteria, eubacteria,

Page 39: A THESIS TITLED - prr.hec.gov.pk

Introduction

12

mammals and plants origin, this family also contains thioglucosidases and 6-

phosphoglycosidases. GH family 1 β-glucosidases are highly active with broad substrate

specificity and also exhibited the activity of β-galactosidase. Approximately 44 β-

glucosidases are belonged to GH family 3, mostly isolated from mold, yeast and some

bacterial source, whereas some hexosaminidases also include in this family. Majority of

fungal β-glucosidases studied and characterized are the members of GH family 3 (Cantarel

et al., 2009; Krisch et al., 2010; Singh et al., 2016). The classification of a family also

demonstrates the glycoside hydrolases evolutionary relationship (Singh et al., 2016).

Generally, β-glucosidases exhibited tremendous diversity in terms of substrate specificity

but little is known about their catalytic mechanism acceptably. Numerous techniques for

example inhibition, pH dependence, essential residues labeling with fluorosugars, isotopic

effect, structure-reactivity studies (with deoxy- substrate analogues) and site-directed

mutagenesis have been applied for elucidating the catalytic behavior and active site

topology of enzyme (Kempton and Withers, 1992; Street et al., 1992; Withers et al., 1992;

Wang et al., 1995). Enzyme hydrolyze the β-glycosidic bonds efficiently between a

polysaccharide and non-polysaccharide moiety.

Most β-glucosidases characterized to date, have been placed in GH family 1 and GH family

3, which are all retaining enzymes, perform their catalytic mechanism in two steps, initially

glycosylation and followed by deglycosylation occurs. Among all reported β-glucosidases,

a conserved glutamic acid (Glu) is the main active site residue (Wang et al., 1995). The

retaining mechanism of β-glucosidase for the catalysis of β-glycosidic bond is completely

demonstrated in figure 1.4. In first step, a glucose-enzyme intermediate complex is formed

by the action of a glutamic acid (Glu) on the anomeric carbon which acts as a nucleophile,

and after that in deglycosylation step, another acid/base catalytic residue activates a water

molecule which hydrolyzes the glycosidic bond to liberate free glucose (monosugar)

molecule (Litzinger et al., 2010).

Page 40: A THESIS TITLED - prr.hec.gov.pk

Introduction

13

Figure 1.4: The catalytic retaining two steps mechanism of β-glucosidase for the hydrolysis of β-glycosidic

bond. Glycosylation, the first step in which a conserved glutamic acid (Glu) residue attacks on the glycosidic

bonds and acts as nucleophile, and by the hydrolytic action of cellobiohydrolase (other enzyme of cellulase

system) cellobiose released. An enzyme-substrate intermediate complex is formed as a result of this. Second

step is known as deglycosylation, by general acid/base catalyst reaction another conserved glutamic acid

(Glu) residue activates a water molecule which further acts on the intermediate complex to release the glucose

molecule.

β-glucosidases are widely found in all form of life and involved in their various essential

bioconversion processes as well as these enzymes have various industrial applications.

Thermotolerant and highly active cellulases, especially β-glucosidases have received

considerable attention, in these days, are extremely desirable to attain a higher degree of

efficiency on the industrial scale and for biotechnological applications. Heat resistant, and

solvent-tolerant enzymes commonly known as ‘extremozymes’ are the valuable tools for

industrial processes in which hardly degradable polymers and compounds need to be

decomposed and liquefied (Elleuche et al., 2014). The quest for hyperstable and acidic-

alkali resistant GHs, mainly cellulolytic enzymes have led the way towards extremophiles

which are adapted to survive at extremely harsh conditions of ecological niches such as

high salt concentrations, extreme of pH, high pressure and temperature. These

microorganisms are the valuable home of unique biocatalysts that can withstand harsh

Page 41: A THESIS TITLED - prr.hec.gov.pk

Introduction

14

conditions of current industrial processes as compare to the prevailing enzymes derived

from mesophiles (mesozymes) (Elleuche et al., 2015).

In recent time, applications and needs of cellulases are completely different with respect to

the various industrial processes such as for biofuel industry prefers thermostable and acidic

resistant enzymes, whereas detergent industry favors enzymes with broad range of pH and

temperature stability as well as halo-tolerant (resistant to salts). However, textile and

industrial wastes treatment industries requires cellulolytic enzymes with heat and alkali

stable characteristics (Kumar et al., 2011). Thermal stability is the most common,

conspicuous and noticeable property of cellulases required in industries. Conducting

biotechnological and industrial processes at elevated temperature has numerous advantages

including higher reaction rate, decrease in viscosity, lower the risk of contamination,

increased solubility of organic compounds (hydrophobic substrates) and improved

bioavailability of hardly degradable environmental pollutants (Niehaus et al., 1999; Kuhad

et al., 2016).

Demand of thermostable β-glucosidases is growing rapidly because of its usage in food

processing and biofuel production. Therefore, the search for enzyme, insensitive to product

inhibition (glucose) with high specific activity and thermal stability, has increased

worldwide. Thermostable β-glucosidases, with high specific activity, have the ability to

function under extreme conditions, a longer hydrolysis time due to stability, lesser amount

used simultaneously and directly along with other heat stable cellulases for saccharification

purpose, as this decreases processing time (by eliminating pre-cooling step where steam is

applied to make biomass more accessible to degradation), improves fermentation yields,

reduces contamination risk, less energy consumption, enhances enzyme activity and

solubility of reactants and products (Liu et al., 2012). Thus, the potential commercial

advantages of efficient and lucrative thermostable cellulases with low degree of inhibition

would reduce the cost of bioethanol production significantly in a simultaneous

saccharification and fermentation (SSF) process (Zhang et al., 2012).

Biomass conversion using cellulases has become a major research area and thermophilic

bacteria are the excellent source of these enzymes (Mehmood et al., 2014). Thermophilic

Page 42: A THESIS TITLED - prr.hec.gov.pk

Introduction

15

fungal cellulases had been extensively studied in last few decades for bioconversion

process (Hong et al., 2007; Yoon et al., 2008; Liu et al., 2012; Ramani et al., 2015; Zhao

et al., 2015). But recently, thermophilic and hyperthermophilic bacteria have accomplished

prodigious consideration due to several reasons such as broad natural diversity, easily

cultivated with cheap nutrients, high growth rate, have great ability for the production of

thermal, alkali and acid-stable cellulases. However, high level of expression, extraction

and purification of bacterial biocatalysts done feasibly as compare to fungal cellulolytic

enzymes (Jabbour et al., 2012; Schroder et al., 2014; Gumerov et al., 2015; Yang et al.,

2015; Chan et al., 2016; Peng et al., 2016).

Hyperthermophiles are the most attractive source of extremely thermotolerant enzymes,

optimally grow at higher temperature usually above 80°C, have been isolated and screened

from different hot deep subterranean locations such as marine sediment, hot springs, oil

reservoir and continental solfatatras. These organisms are thought to be the least evolved,

have deeper phylogenetic branches, most of them belong to archaea domain while some

are the members of genera Thermotoga and Aquifex in bacterial domain (Huber et al., 2000;

Takahata et al., 2001). Certain bacterial species across different genus have been reported

to produce highly thermostable cellulases including Aquifex, Alicyclobacillus,

Fervidobacterium, Geobacillus, Thermoanaerobacterium, Caldicellulosiruptor,

Dictyoglomus, Rhodothermus, Anoxybacillus and Thermotoga.

During the last few years many biocatalysts have been successfully isolated from these

exotic microbes which has exceptional features such as resistant to extreme pH,

temperature and chemicals denaturants (organic solvents, detergents and chaotropic

agents). Hence, these biocatalysts can be used as a model for constructing and designing

proteins with unique properties for industrial and biotechnological applications (Barnard

et al., 2010; Chang and Yao, 2011; Jager et al., 2012; Bhalla et al., 2013; Chen et al.,

2013). Only Less than 5% of hyperthermophilic archaea and bacteria are cultivable in the

laboratory due to their unique and exceptional cultivation requirements (Elleuche et al.,

2015). Therefore, to get the enhanced production of heat loving cellulolytic enzymes

particularly β-glucosidases, various approaches have been adopted by researchers in which

the most prominent strategy is cloning of thermophilic genes in suitable mesophilic hosts

Page 43: A THESIS TITLED - prr.hec.gov.pk

Introduction

16

(Bacillus spp., Escherichia coli, Aspergillus sp., Saccharomyces cerevisiae, and Pichia

pastoris) using efficient expression vectors. Mostly, thermophilic heterologous proteins

produced in mesophiles are not hydrolyzed by host proteases; correctly and efficiently

folded at low (37-40°C) temperature, able to maintain their hydrolytic activity and stability,

and can be purified conveniently by using thermal denaturation (precipitation) of host

proteins. Usually, the degree of obtained protein purity is adequate and appropriate for

several industrial applications (Niehaus et al., 1999).

Cloned β-glucosidases exhibited prodigious advantages including hydrolyzing ability of

isoflavone (Kuo and Lee, 2007), maximum expression, thermo-active and more resistant

to end product inhibition (Jabbour et al., 2012). Various mesophilic host system were used

for achieving high level of heterologous protein expression but E. coli species along with

pET expression vectors series has been proved an excellent and well-established expression

platform (Studier, 2005). Some bacterial thermostable β-glucosidases that showed glucose

tolerant behavior have been cloned previously from Thermoanaerobacterium

thermosaccharolyticum (Pei et al., 2012), Fervidobacterium islandicum (Jabbour et al.,

2012), Caldicellulosiruptor bescii (Bai et al., 2013), Acidilobus saccharovorans (Gumerov

et al., 2015) Thermoanaerobacterium aotearoense (Yang et al., 2015) and overexpressed

in E. coli host. However, these β-glucosidases are not as much glucose resistant as

exceedingly required for biomass degradation and there are limited reports on kinetic and

thermodynamics study of β-glucosidases.

The members of genus Thermotoga has distinct features from other bacteria, optimally

grow at 80-90°C, they are obligate anaerobes, fermentative heterotrophs, rod-shaped,

gram-negative, non-spore forming cells with an outer prominent sheath-like structure or an

envelope known as ‘toga’ and having balloon like structure at both ends (Figure 1.5a). The

presence of sheath ‘toga’ is most noticeable characteristic of all the members of order

Thermotogales, include Thermotoga neapolitana, Thermotoga sp. strain RQ7, Thermotoga

sp. strain FjSS3-B.1, Thermotoga subterranea, Thermotoga elfii, Thermotoga thermarum,

Thermotoga hypogea, Thermotoga maritima and Thermotoga lettingae, have been found

naturally in diverse geographical niches (Figure 1.5b), while two novel species of genus

Thermotoga isolated from the Kubiki oil reservoir from Japan, which called as Thermotoga

Page 44: A THESIS TITLED - prr.hec.gov.pk

Introduction

17

petrophila RKU-1T and Thermotoga naphthophila RKU-10T. The genomic sequence and

phenotypic characteristics revealed that both T. petrophila and T. naphthophila have great

similarity with T. neapolitana and T. maritima (Takahata et al., 2001). The species belong

to genus Thermotoga have the ability to degrade simple and complex sugars effectively,

and are believed to be an excellent source of highly active, resistant to denaturing agents

and heat-stable GHs specifically cellulolytic enzymes (Park et al., 2005; Turner et al.,

2007; Xue et al., 2009; Zhao et al., 2013; Mahmood et al., 2014; Xie et al., 2015; Long et

al., 2016).

Figure 1.5: (a) General structure of Thermotoga bacterium. (b) Octopus Spring, water from the main

spring supports the growth of a variety of Thermotoga species.

Figure 1.6: Electron micrographs of stained ultrathin section of Thermotoga naphthophila RKU-10T strain.

Arrows indicate the cell wall (cw) and toga (t) (Takahata et al., 2001).

(a) (b)

Page 45: A THESIS TITLED - prr.hec.gov.pk

Introduction

18

Thermotoga naphthophila (naphthophila is a Greek word means bitumen-loving), a marine

hyperthermophilic eubacterium has all the general characteristic of family

Thermotogaceae such as obligate anaerobe, rod-shaped and non-spore-forming. Optimally

grow at pH 7.0 and 80°C temperature, cells are 0.8-1.2 µm wide and 02-07 µm long,

heterotrophic bacterial cell of T. naphthophila has a notable outer sheath ‘toga’ with several

lateral and subpolar flagella (Figure 1.6). Whereas, T. neapolitana lacks flagella and a

single sub-polar flagellum present in T. maritima (Takahata et al., 2001). Thermotoga

naphthophila RKU-10T (ATCC BAA-489 / DSM 13996 / JCM 10882) contains a large

number of putative cellulolytic genes.

Therefore, in this present research work two hyperthermophilic novel cellulolytic genes of

T. naphthophila are cloned and overexpressed in E. coli BL21 CodonPlus (DE3)-RIPL, a

mesophilic expression host. Various cultivation and induction strategies have been

developed for enhancing the production of engineered E. coli BL21 CodonPlus (DE3)-

RIPL cells and attaining high-level expression of both enzymes. Heat treatment followed

by chromatographic techniques are applied for the purification of thermo-efficient cloned

enzymes. Biochemical characterization of recombinant β-1,4-glucosidases together with

kinetic and thermodynamic analysis is done which give insights about the thermostability

of enzymes. Industrially, they could effectively use for biodegradation of carbohydrates,

moreover this study has proved that highly thermophilic bacteria are the

impending resources for isolating extremely effective glycoside hydrolases for

biochemical and biofuel production.

Page 46: A THESIS TITLED - prr.hec.gov.pk

Introduction

19

1.2. Objectives

The growing demands of biofuel (bioenergy) has led to the emphasis on novel and highly

active cellulolytic enzymes to improve efficiency and rate of bioconversion process.

Therefore, the search of novel highly active thermostable cellulolytic enzymes is the great

need of time but the cultivation conditions of hyperthermophiles are highly demanding.

Hence, with the help of recombinant DNA technology, it is possible to achieve their

hyperstable enzymes in mesophiles; cloning of hyperthermophilic cellulolytic genes into

heterologous mesophilic host provides an effective approach to accomplish the high level

of enzyme expression. Furthermore, cloning of individual gene helps to study the

biochemical characterization of that recombinant enzyme.

The present research work is intended to clone two highly thermostable cellulolytic β-

glucosidase genes from T. naphthophila and overexpress in mesophilic host. Purify the

cloned β-glucosidases to evaluate their properties by complete characterization. The major

goal of this study is to produce potent hyperthermostable β-glucosidase with high catalytic

activity and low product inhibition. The following steps are followed to achieve the aim of

present study:

Amplification of cellulolytic β-glucosidase genes from T. naphthophila.

TA cloning of amplified product of β-glucosidase genes.

Sequencing of genes

Sub-cloning in expression vector.

Expression of heterologous proteins in E. coli BL21 CodonPlus DE3-(RIPL).

Optimization of cultivation conditions.

Analysis of recombinant proteins expression by enzymatic assays and SDS-PAGE.

Purification of β-glucosidases using chromatographic techniques.

Characterization of cloned β-glucosidases.

Kinetic and thermodynamic analysis of proteins.

Page 47: A THESIS TITLED - prr.hec.gov.pk

CHAPTER-II

REVIEW OF LITERATURE

Page 48: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

20

2. Review of Literature

Bioethanol has been widely accepted to be the most appropriate replacement of

petroleum derived from fossil fuels, hence the rapidly increasing demands of bioethanol

has directed to the importance of active, competent, novel and thermo-efficient

cellulolytic enzymes to enhance the productivity of bioconversion process of plant

biomass. During last few years, many cellulolytic genes have been identified, isolated,

expressed, purified and characterized but the appropriate and impervious cellulases have

not been found yet, which can tolerate all biological challenges of the bio-

saccharification process. Therefore, recombinant thermophilic and hyperthermophilic

bacterial cellulolytic biocatalysts that overexpressed in mesophilic hosts, are the most

suitable candidates, because of their tolerance and resistance to the harsh conditions of

biodegradation process. Thermostable cellulases, especially β-glucosidases, which have

the ability to performed catalytic activity under extreme conditions, highly stable, and

effectively used in various cocktail with other cellulases, as this enhances the yields and

decreases the energy cost of the process (Liu et al., 2012).

In the following section, various aspect of thermo-activated beta-glucosidases relating to

cloning, recombinant protein production, purification, structure analysis, characterization

and applications have been reviewed.

Gabelsberger et al. (1993) identified, isolated and cloned a hyperthermophilic β-

glucosidase from Thermotoga maritima. The recombinant enzyme was overexpressed

and purified from a mesophilic host (Escherichia coli). By size exclusion

chromatography, the apparent molecular weight (MW) of dimer enzyme almost 95 kDa

was determined. Enzyme consisted of two similar subunits of 47 kDa, was determined

by SDS-PAGE. Purified β-glucosidase had great specificity of substrate and

commendably hydrolyzed β-glucoside, followed by β-fucoside, β-galactoside, and β-

xyloside substrates. Kinetic parameters for the hydrolysis of 0.05–20 mM oNPG and

0.1–10 mM oNPGal at 75°C, a non-linear Lineweaver-Burk formed. Lactose and

cellobiose did not cause any positive influence on enzyme activity, while 350 mM

lactose inhibited it completely. Enzyme was optimally hydrolyzed pNPG at pH 6.1. It

Page 49: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

21

exhibited high thermo-tolerant behavior, and pure β-glucosidase (50 μg mL-1) retained

60% residual activity without the addition of any additive dissolved in buffer of pH 6.2,

after being the incubated for 360 min at 95°C.

Kengen et al. (1993) revealed that a hyperthermophilic and highly active β-glucosidase

was found in the cell-free extract of Pyrococcus furiosus with high specific activity. The

crude enzyme was purified to homogeneity with the apparent MW of 58 kDa, has 4.40 pI

value; and characterized completely. Enzyme showed its peak catalytic activity with 446

U mg-1 specific activity at 102 to 105°C and at slightly acidic pH 5.0, against the

substrate of pNPG. Purified BglA activity did not depend on thiol groups, nor on high ion

strength and divalent cations. It exhibited affinity with pNPG, pNPGal, pNPX and

cellobiose substrates, while showed no hydrolytic activity against disaccharides or

polysaccharides like cellulose. Kinetic parameters were studies for pNPG and cellobiose

using Lineweaver-Burk plots and determined Km of 0.15 mM and 20 mM, Vmax of 700 U

mg-1 and 470 U mg-1 for pNPG and cellobiose, respectively. Purified enzyme had

remarkable thermal stability with a half-life of 780 minutes at 100°C.

Ruttersmith and Daniel (1993) isolated a thermostable beta-glucosidase gene from a

hyperthermophilic bacterium Thermotoga sp. strain FjSS3-B.1, cultivated at 80°C, and

after production of enzyme, purified to homogeneity using different strategies like anion

exchange followed by chromatofocusing and gel filtration. On SDS-PAGE, a single band

of purified enzyme with a MW of 75 kDa was observed. Enzyme had an optimal activity

of 195 U mg-1 at pH 7.0 and 80°C temperature. Enzyme had broad substrate specificity

and best catalytic activity was found towards pNPG substrate, with Km value of 0.1 mM.

Purified enzyme activity was inhibited by glucose (Ki value of 0.42 mM), and did not

effect by 40 mM xylose. Purified beta-glucosidase did not show any influence or

inhibitory effect in the presence of metal ions in fact, Ag2+ and Co2+ stimulated the

catalytic efficiency of the enzyme, only Cd2+, Ni2+ and Hg2+ obviously repressed the

activity. DTT, ethanol and Triton X–100 highly enhanced the activity of the beta-

glucosidase. Purified enzyme was tremendously thermotolerant with a half-life of 150

min at 98°C temperature and its stability improved in the presence of betaine or trehalose.

Page 50: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

22

According to Voorhorst et al. (1995) a hyperthermophilic cellobiose-hydrolyzing

biocatalyst known as beta-glucosidase (CelB) was cloned from Pyrococcus furiosus and

overexpressed in prokaryotic host E. coli. Deduced CelB sequence of amino acid had

great similarity with beta-glycosidases that were the members of glycosyl hydrolase (GH)

family 1. High-level production of beta-glucosidase (20% from total cell protein) was

successfully purified by two-step method that included thermoincubation for 40 min at

80° followed by anion-exchange chromatography. Purified enzyme had a MW of 58 kDa

estimated by gel electrophoresis (SDS-PAGE). Purified enzyme displayed peak activity

at 105°C and slightly acidic pH 5.0 against pNPG substrate. Km of 0.41 mM and Vmax of

1,169 µmol min-1 mg-1 was determined using pNPG.

Breves et al. (1997) sequenced a genomic DNA fragment of 5.9 kb from a thermophilic

anaerobe Thermoanaerobacter brockii. The DNA fragment had two genes, which located

adjacent to each other. The two different beta-glucosidases, XglS and CglT, belonged to

different glycosyl hydrolases (GHs) families with MW of 81 kDa and 52 kDa,

respectively. Both genes were overexpressed in E. coli, and recombinant CglT was

recognized by N-terminal sequencing and purified by heat denaturation and fast protein

liquid chromatography, after purification MW of 50 kDa was detected by gel

electrophoresis. Enzyme CglT had peak activity with pNPG, at optimal pH of 5.5 and at

75°C temperature. CglT exhibited thermostability for 24 h at 60°C. Enzyme CglT had the

ability to hydrolyzed various disaccharides as well as β-1,4-cello oligomers. CglT had

affinity for pNPGal and pNPF, while XglS cleaved both β-xylosides and β-glucosides as

well. Glucose moderately inhibited the enzyme activity and the Ki value for glucose was

determined to 200 mM.

Zverlov et al. (1997) cloned and sequenced a thermo-tolerant beta-glucosidase (BglB)

from the genomic DNA of Thermotoga neapolitana. Amino acid sequence alignment

showed that the cloned BglB enzyme was a candidate of GH family 3 and belonged to the

class EC 3.2.1.21. The gene was sub-cloned in pTT51 vector, and followed by

overexpressed in E. coli, and completely purified by thermal precipitation of heat labial

host proteins afterward ion exchange (DEAE column) and hydrophobic interaction

chromatography. SDS-PAGE analysis of purified fraction revealed one sharp band of

Page 51: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

23

protein with a monomeric MW of 81 kDa. Purified BglB had 255 U mg-1 specific activity

on pNPG at the optima of pH (5.5) and temperature (90°C). Km values for cellobiose,

laminaibiose and pNPG were 50, 10 and 0.1 mM, respectively.

Li and Lee (1999) studied gene cloning and expression of a beta-glucosidase from

Flavobacterium meningosepticum, which contained 2,178 bp encoded a protein of 726

amino acids. On the base of sequence analysis, the enzyme was classified as a GH3

family member. The recombinant E. coli cells were induced with IPTG, cultivated in LB-

medium, the high-level expression of enzyme was achieved after 6 hours induction. The

cloned beta-glucosidase enzyme was purified using cation-exchange chromatography, the

purity and 79.95 kDa molecular mass was determined by SDS-PAGE. Optimal

temperature and pH to achieved maximum activity of cloned enzyme (2,369 U mg-1) was

50°C and 5.0, respectively. It was remained stable at pH 5.0–8.1 (25°C temperature).

Recombinant enzyme effectively hydrolyzed aryl-beta-D-glycosides whereas, pNPG was

the most preferred substrate, with Km value of 0.68 mM.

Goyal et al. (2001) cloned, overexpressed, purified and characterized a β-glucosidase

(BglB) of a hyperthermophilic eubacterium Thermotoga maritima in an expression host

E. coli to get high level of heterologous active protein. Heat treatment and ion exchange

chromatography techniques were applied to purify recombinant BglB enzyme, and the

final fraction was subjected to analysis of SDS-PAGE which showed a single band of 81

kDa molecular mass. Optimal temperature and pH at which BglB displayed maximum

hydrolytic activity was 85°C and 5.0, respectively whereas at acidic pH (3.5) and lower

temperature (70°C) enzyme also showed high catalytic activity. BglB demonstrated a

broad range of pH stability from 5.0 to 9.0, and retained 68% residual activity at pH 10.

Enzyme exhibited thermo-tolerant behavior and remained stable up to 75°C temperature

for 30 min. BglB had a broad substrate specificity and showed great affinity towards

pNPG substrate with kcat and Km values of 6.34 s−1 and 0.0039 mM, respectively. Purified

BglB hydrolytic activity was activated in the presence of organic compounds and straight

chain alcohols.

Page 52: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

24

Parry et al. (2001) reported a thermo-tolerant extracellular, active and soluble β-

glucosidase enzyme from Thermoascus aurantiacus was purified using ion exchange and

Mono-P column chromatography techniques. The purified enzyme was homogeneous

with a MW of approximately 120 kDa, presented optimal catalytic activity at 80°C

temperature and pH 4.5, however at 90°C β-glucosidase displayed 70% relative activity.

Purified enzyme had high thermal and pH stability, at 70°C (pH 5.2) β-glucosidase

remained stable up to 48 h while above and below 70°C and pH 5.0 enzyme stability

decreased. It had the ability to hydrolyzed substrates efficiently with a β-glycosidic

linkage, cello-oligosaccharides, alkyl and aryl β-D-glucosides. Kinetic studies

demonstrated the lowest Km value of 0.1137 mM for pNPG and maximum kcat value of

17052 min-1 towards cellobiose. Straight chain alcohol activated the β-glucosidase while

glucose and D-δ-gluconolactone competitively repressed the activity with Ki values of

0.29 mM and 8.3 mM, respectively. Transglycosylation and hydrolytic activity was

optimally done in an acidic environment although enzyme catalytic behavior was

enhanced at high concentration of alcohols. The stereochemistry analysis of cellobiose

hydrolysis showed that purified β-glucosidase was a retaining glycosidase, and the

sequence alignment of N-terminal amino acid specified that β-glucosidase from

Thermoascus aurantiacus belonged to GH family 3.

Molecular cloning of a thermo-tolerant gene of beta-glycosidase with 1,311 bp encoding

437 amino acids from a hyperthermostable bacterium Thermus nonproteolyticus HG102

was carried out by Xiangyuan and co-worker (2001). Enzyme was overexpressed in E.

coli host and purified to homogeneity with a MW of 48,900 Da on SDS-PAGE. The

monomeric enzyme exhibited peak activity at 90 degrees C and at 5.6 pH, with high

catalytic activity towards pNPG. It can also effectively hydrolyzed pNPGal, pNPF and

pNPMan substrates. The kinetic parameters exhibited that the kcat/Km ratio for pNPG and

pNPF was higher than for pNPMan. The recombinant enzyme showed high

thermostability at 90 degrees C with a half-life of 2.5 hours, and displayed pH stability

over a wide range.

Zanoelo et al. (2004) characterized an inducible β-glucosidase enzyme from a Scytalidum

thermophilum a thermophilic fungus. The enzyme had pI value of 6.5 and purified by gel

Page 53: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

25

filtration and ion-exchange chromatography, and the final fraction exhibited a single

protein band of 40 kDa. Optima of temperature and pH of purified enzyme were 60°C

and 6.5, respectively against pNPG substrate. It exhibited thermal stability at 50–55°C,

with a 20 min half-life at 55°C temperature. Purified enzyme hydrolyzed various

substrates such as cellobiose, lactose, pNPX, and oNPGal whereas maximum affinity was

observed with pNPG. Km values for cellobiose and pNPG were 1.61 mM and 0.29 mM,

however Vmax for cellobiose and pNPG were 4.12 U mg-1 and 13.27 U mg-1, respectively.

The activity of enzyme was stimulated by the addition of 50 to 200 mM xylose and

glucose concentration and the Ki values of 43.24 mM for xylose and 36.69 mM for

glucose.

Kang et al. (2005) identified and cloned three thermo-tolerant genes in which a β-

galactosidase (bgaA) and two β-glycosidases (bglA and bglB) from a heat loving

bacterium Thermus sp. IB-21. The genes bgaA, bglA and bglB contained 1,938 bp, 1,311

bp and 1,296 bp of nucleotides which encoded 645, 436 and 431 amino acid residues,

respectively. These three genes were sub-cloned in pET-21b(+) expression vector,

propagated and overexpressed in E. coli BL21 (DE3). Heat treatment at 70°C for 40 min

followed by affinity chromatography techniques were applied for the purification of all

recombinant enzymes. Enzymes homogeneity and purity was analyzed by SDS-PAGE.

All purified recombinant enzymes exhibited maximum activity at pH 5.0–6.0.

Park et al. (2005) reported a molecular gene cloning of a hyperthermotolerant beta-

glucosidase (bglA) of 1,350 bp from Thermotoga neapolitana. The bglA gene was sub-

cloned in p6xHis119 vector, and subsequently overexpressed in E. coli. To purify cloned

BglA, crude supernatant was subjected to heat-treatment at 75°C to remove heat-labile

proteins followed by Ni-NTA affinity column chromatography. SDS-Page analysis

exhibited that purified TnBglA had 52 kDa protein. At 95°C recombinant purified BglA

showed peak catalytic activity and enzyme had great thermal stability with a half-life of

12 min at 105°C. TnBglA was optimally active over a range of 5.0–7.0 pH and remained

stable between 4.5–7.5 pH. TnBglA had high substrate specificity towards natural

(lactose and cellobiose) and artificial substrates (pNPG). BglA kinetic studies revealed

the values of Km (0.89 mM), kcat (657.2 s-1) and kcat Km-1 (753.91 mM-1 s-1) for pNPG

Page 54: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

26

substrate. Chemical inhibitors and metallic cations did not significantly influence the

activity of the recombinant enzyme, whereas beta-mercaptoethanol and dithiothreitol

markedly enhanced BglA enzymatic activity.

Hong et al. (2006) cloned a hyperthermophilic β-glucosidase gene (BglB) of 2.9 kb from

a anaerobic bacterium Thermotoga maritima MSB8 into pET-21a(+)vector and

heterologously overexpressed in E. coli BL21. Sequence homology of BglB with other β-

glucosidases from variety of organisms revealed that BglB was a conspicuous member of

GH family 3. After the enzyme inducement and production, crude BglB was purified by

applying affinity column chromatography followed by final eluted protein with a single

band of 81 kDa molecular mass was analyzed on native and SDS-PAGE. Purified BglB

hydrolyzed several substrates effectively such as pNPG, salicin, and arbutin, but peak

activity was found with pNPG substrate at 80°C temperature and 7.0 pH. Purified BglB

remained stable at high temperature of 80°C for up to 7 hours. The divalent cation Hg2+,

Cu2+, and Zn2+ repressed the enzyme activity.

In 2007, Hong and co-worker isolated, cloned and sequenced a thermo-tolerant beta-

glucosidase gene (bgl1) of Thermoascus aurantiacus IFO9748, and overexpressed in

yeast Pichia pastoris. Cloned gene consisted of 2,828 bp and deduced sequence of amino

acids displayed high homology with others GH family 3 glucosidases which confirmed

that this enzyme belonged to family 3. The inducible, active and recombinant enzyme

Bgl1 was purified to homogeneity and subjected to complete biochemical

characterizations. The purified enzyme showed maximum specific activity of 23.3 U mg-1

at 70 degree celsius and pH 5.0 using pNPG as a substate. Bgl1 retained more than 70%

residual activity after 60 minutes incubation at 60°C. Purified enzyme had affinity with

various artificial substrates such as pNPX, pNPC as well as cello-oligosaccharides and

cellobiose. Kinetic parameters were studies for pNPG, pNPX, pNPC and cellobiose. The

recombinant Bgl1 had the ability to utilized cellobiose as a source of carbon.

Kim et al. (2007) isolated a novel β-glucosidase gene (bglA) from uncultured soil

bacteria during the construction of genomic libraries from soil. Amplified product was

cloned in pET-32a(+) vector followed by heterologously expressed in E. coli BL-21

Page 55: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

27

(DE3) host cells. The deduced molecular mass of bglA amino acid sequence was 55-kDa,

which showed 56% similarity with a β-glucosidase of Chloroflexus aurantiacus belonged

to glycosyl hydrolase family 1. The crude extract was purified by applying Ni-NTA

agarose slurry and exhibited optimal pNPG degradation activity of purified enzyme was

observed at fifty five degree celsius and 6.5 pH. Enzyme showed thermal and pH

stability, retained 82% residual activity at 50°C after 60 minutes incubation and broadly

stable at pH 5.5 to 9.0. Its activity was enhanced in the presence of Mn2+, Mg2+, and Cu2+

divalent cations, while Co2+, Zu2+, Ca2+, and Fe2+ had negative effects on enzyme

activity. The purified enzyme had Km and Vmax values using pNPG substrate were 0.16

mM and 19.10 µmol min-1, respectively. Furthermore, purified enzyme displayed broad

substrates specificity and hydrolyzed both natural and synthetic substrate easily.

Kaur et al. (2007) purified and characterized a thermophilic, soluble and active β-

glucosidase from Melanocarpus sp. Enzyme had pI of 4.1, using DEAE-Sepharose (FF)

and Poly-buffer exchanger (PBE 94) column chromatography were used for the

purification of enzyme, 92 kDa molecular mass was determined on SDS-PAGE analysis.

Purified enzyme exhibited activity of 10.04 U mg-1 at the optimal assay conditions 60°C

temperature and pH 6.0. Presence of metallic cations, mercaptoethanol and DDT

influenced the catalytic efficiency of enzyme positively while only Cu2+ repressed the

activity up to 38 %. Enzyme hydrolytic efficiency was evaluated using various substrates,

and found pNPG as the most preferred substrate and low affinity for pNPC. Km (3.3 mM)

and Vmax (43.68 µmolmin-1mg protein-1) of purified β-glucosidase was determined for the

hydrolysis of pNPG, and kcat value of 4×103 min-1 was quantified.

Park et al. 2007 cloned and overexpressed a hyperthermostable beta-glycosidase of an

archaeon, Sulfolobus shibatae in E. coli BL21 (DE3) cells using pRSET-B expression

vector. Maximum expression of enzyme was observed after inducing recombinant

bacterial culture with 1 mM IPTG after that incubated for 3 h at 37°C. The heterologous

protein was efficiently purified using two-step method including heat treatment for 30

min at 70°C followed by affinity chromatography technique. The analysis on SDS-PAGE

demonstrated that enzyme was homogeneous with 57 kDa molecular mass. Purified

enzyme showed activity maximum at pH 5.0 buffer with pNPG substrate and at 95°C.

Page 56: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

28

Enzyme displayed high thermal stability between 70 to 100°C, and had a half-life of 900

minutes at temperature 75°C. It showed broad substrate specificity, mostly metal ions did

not affect the enzyme activity, though 10 mM MnCl2 stimulated its catalytic efficiency.

2-mercaptoethanol and dithiothreitol (DTT) chemicals significantly enhanced the

hydrolysis activity of enzyme.

According to Turner et al. (2007) a novel highly thermo-tolerant gene of β-glucosidase

from Thermotoga neapolitana was cloned and overexpressed in E. coli. Open reading

frame of gene encoded a 721 amino acid residues, the sequence alignment confirmed that

β-glucosidase (TnBgl3B) was a member of glycosyl hydrolase family 3 to be a competent

enzyme in alkyl-glucoside reactions with alcohol as the acceptor molecule using

transglycosylation. Recombinant E. coli cells were induced with IPTG, overexpressed,

active and soluble TnBgl3B was purified to homogeneity with 81 kDa MW using heat

treatment and affinity chromatography. At 90°C and pH 5.6, purified TnBgl3B was

optimally active while alcoholysis/hydrolysis ratio more favorable at 60°C (lower

temperature) where total activity was dropped, however water content and pH were more

heavy effected this ratio. Kinetic study for Km and Vmax were determined 0.11+0.03 mM

and 93+13 U mg-1, using pNPG substrate, respectively.

Kuo and Lee, (2008) studied the genomic sequence of a strain called

Bacillus subtilis natto, on the basis of genomic study two beta-glucosidase genes (yckE

and bglH) were isolated, cloned in E. coli M15 strain and overexpressed after the

induction with IPTG. The recombinant enzymes were active, soluble and highly

expressed in host cells, the enzymes were purified using affinity chromatography. The

YckE and BglH enzymes analyzed by SDS-PAGE which were approximately 54 kDa and

53 kDa, respectively. Recombinant YckE and BglH showed maximum hydrolyzing

activity towards pNPG substrate at 37–45°C, and enzymes displayed thermal stability up

to 45°C, while BglH had more heat stability than YckE enzyme. Both purified enzymes

highly active at pH 6.0, both enzymes fairly stable over a broad pH range, while BglH

was less stable than YckE. By the addition of divalent metallic cations (1 mM) the

enzymes activity were inhibited by only 0-23%, while Cu2+, Fe2+ and Cd2+ ions repressed

the recombinant BglH activity by 43%, 63% and 73%, whereas the activity of YckE was

Page 57: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

29

moderately repressed (not more than 20%) in the presence of any divalent metal cations.

YckE exhibited great affinity towards pNPF as compared to pNPG, and BglH revealed the

maximum affinity for genistin. The catalytic efficiency (kcat Km-1) of BglH for pNPG,

pNPF and genistin were 3.01, 0.90 and 147.20 mM-1 s-1, respectively. The kcat Km-1 of

YckE for pNPG, pNPF and genistin were 2.44, 306.57 and 19.45 mM-1 s-1, respectively.

Yoon et al. (2008) reported an extracellular and highly active beta-glucosidase enzyme

from Fomitopsis palustris was produced and purified to electrophoretic homogeneity.

The molecular mass of purified beta-glucosidase was determined by using SDS-PAGE,

which displayed a single band of protein to be approximately 138 kDa. Amino acid

sequence analysis was determined that suggested beta-glucosidase from Fomitopsis

palustris had homology with glycosyl hydrolase (GH) family 3 therefore, belonged to

GH3. Purified enzyme displayed prime activity toward pNPG at 70 degrees and pH 4.5.

Enzyme indicated thermal stability, with 97 h and 15 h half-life at 55°C and 65°C,

respectively. The Km value for cellobiose (4.81 mM) and pNPG (0.117 mM) were

calculated along with kcat values for cellobiose and pNPG were 101.8 and 721 sec-1,

respectively. In the presence of gluconolactone and glucose, the purified beta-glucosidase

catalytic efficiency was competitively inhibited and showed Ki values of 0.008 mM and

0.35 mM, respectively using pNPG as a substrate. Enzyme showed high affinity with

cellobiose and pNPG but had no activity toward CMC and xylan substrates.

Bajaj et al. (2009) reported a study on isolation and characterization of β-glucosidase

from a bacterial strain named M+. Enzyme was purified to homogeneity using ion

exchange chromatography, and SDS-PAGE displayed a single band of purified β-

glucosidase with 24 kDa molecular mass. Optimal enzyme activity (2200 U L-1) was

found against pNPG substrate at 50°C temperature in acetate buffer of pH 6.0, while β-

glucosidase showed considerably good catalytic activity at pH 5.0, 7.0–9.0 and also

exhibited good catalytic activity at 60–80°C temperature with 1600–1900 U L-1. Purified

β-glucosidase displayed thermo-tolerant behavior, and was thoroughly active at broad

range of temperature 50–80°C and retained 75–100% activity after 60 min incubation.

Enzyme also revealed pH stability over a range of 5.0–9.0. Enzyme exposed resistance to

Page 58: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

30

the metal ions and chemical additives while in the presence of Pb2+ enzyme catalytic

behavior boost up 47%.

A hyperthermophilic bacterium producing beta-glycosidase an extracellular active

enzyme, was isolated from hot springs of Japan, and cultured in laboratory at optimal

growth conditions (temperature 80°C and pH 6.5) for 48 h, was carried out by Gu et al.

(2009). The bacterium belonged to genus Thermus, found out after morphological study

and sequencing, bacterium was Thermus thermophilus HJ6 strain with highest cultivation

temperature as compared to others members of genus Thermus. The genomic DNA was

used as template for getting the multiple copies of beta-glycosidase gene with 1,296 bp,

which encoding a protein (431 amino acids) with a predicted MW of 48.7 kDa. The gene

was cloned, sequenced and highly expressed recombinant protein was purified to

homogeneity from the host cells of E. coli BL21 CodonPlus. The heterologous protein

purity was confirmed by SDS-PAGE, which had a single band of 45 kDa. Purified

enzyme showed best activity at pH 8.5 and temperature 90°C with pNPG substrate.

Enzyme had showed great thermal tolerance, with a 30 min half-life of thermal

inactivation at 95°C. Recombinant enzyme displayed Km, kcat and catalytic efficiency kcat

Km-1 values for pNPG was 0.16 mM, 9.24 s-1 and 57.12 mM-1 s-1, respectively. Kinetic

study revealed that pNPF and pNPG substrates were better than the pNPGal. The kcat Km-1

of recombinant enzyme was 70-fold increased at 80°C to that 40°C, which indicated that

high temperature activated the enzyme catalytic efficiency.

Harnpicharnchai et al. (2009) demonstrated the isolation of an endophytic fungal strain,

Periconia sp. BCC2871 for the production of thermo-efficient beta-glucosidase (BGLI).

The complete gene of beta-glucosidase was identified, expressed in the host strain Pichia

pastoris KM71. High-level of expressed heterologous protein was purified to

homogeneity, the purified recombinant enzyme showed the similar biochemical behavior

as the native beta-glucosidase, optimal enzyme activity at 70°C temperature and pH of

5.0–6.0 was observed. Cloned enzyme remained stable even after the incubation at high

temperature for long time period, 60% relative activity was found after 90 min incubation

at 70°C. Purified enzyme was quite stable under neutral to basic conditions, and retained

100% enzyme activity at pH 8.0 after being incubated for 120 min. Heat inactivation of

Page 59: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

31

enzyme was enhanced remarkably by the addition of sucrose, glucose and cellobiose.

Purified BGLI had maximum affinity towards synthetic substrates having glycosyl

groups, cellobiose and CMC.

According to Hong et al. (2009) a thermo-efficient β-glucosidase gene isolated and

cloned from a thermophilic bacterium Caldicellulosiruptor saccharolyticus DSM 8903,

and was expressed effectively in host cells of E. coli. Heterologous enzyme was

efficaciously purified to homogeneity using heat treatment followed by His-Trap affinity

chromatography, and final fraction presented a single band of 54 kDa on gel. Optimal

temperature 70°C and pH 5.5 of enzyme was found with pNPG substrate. Purified β-

glucosidase showed great thermal stability at 60–80°C, with half-lives of 250, 24.3, and

0.4 hours at 60, 70, and 80°C, respectively. The enzyme exposed no catalytic activity

against aryl-alpha-glycosides while had great affinity towards aryl-beta-glycosides such

as pNPF, pNPG, and pNPGal in decreasing order.

Kim et al. (2009) isolated the genomic DNA of Thermoplasma acidophilum, which

employed as source of bglA gene, a putative β-galactosidase. The bglA gene (1,452 bp)

cloned, expressed, purified and characterized in host E. coli ER2566 strain. The sequence

analysis of amino acid verified that the putative enzyme was a member of GH family 1.

The recombinant enzyme was purified with a single protein band of 57 kDa on SDS-

PAGE and had 82 U mg-1 specific activity by using pNPG. Enzyme maximum catalytic

activity was found at pH 6.0 and 90°C; recombinant enzyme showed high thermal

stability in pH 6.0 buffer, with 0.4 h half-life at 90°C while it displayed less thermostable

behavior below pH 6.0 and showed only 13 h stability at 75°C. Enzyme activity did not

activated or influenced by mono- and divalent metal cations and not effected by EDTA

chelating agent. Recombinant enzyme showed high substrate specificity, and exhibited

maximum towards pNPF and pNPG, with km value of 0.2 mM and 0.4 mM, kcat value of

182 s-1 and 141 s-1, and kcat Km-1 value of 381 s-1 mM-1 and 1,068 s-1 mM-1, respectively.

In 2009, Xue and co-workers reported cloning and overexpression of a hyperthermostable

beta-glycosidase A gene (bglA) from Thermotoga maritima in E. coli BL21 CodonPlus

using pET-20b(+) vector. The expression of soluble heterologous protein (BglA) was

Page 60: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

32

enhanced from 12.9–15.4% in total soluble cell proteins at 16°C temperature of

induction with low concentration of IPTG inducer. The purified BglA enzyme exhibited

66.8 U mg-1 specific activity at the optimal temperature of 90°C and pH 6.2. BglA

showed stability from pH 4.2–8.2 and heat stability retained almost 90% residual

activity at 90°C after 60 min incubation. Purified BglA exhibited great affinity with

pNPG substrate with Km, kcat Km-1 and Vmax values of 0.43 mM, 589.5 mM-1 s-1 and

323.6 U mg−1, respectively while Km (9.0 mM), kcat Km-1 (16 mM-1 s-1) and Vmax

(183.2 U mg−1) values were found against salicin.

Fang et al. 2010 reported a beta-glucosidase gene (bgl1A) encoded an active protein of

442 amino acids residues, which was isolated and screened from a microbial

metagenomic library of marine. The sequence analysis of Bgl1A protein showed that

enzyme was a member of GH family 1. The gene bgl1A was ligated in pET22b(+) and

transformed into E. coli BL21 (DE3) and recombinant Bgl1A showed high level of

expression in LB-medium after 7 h isopropyl-β-D-thiogalactoside (IPTG) induction. The

enzyme was purified using affinity chromatography with the MW of 51 kDa by SDS-

PAGE analysis. Bgl1A showed great stability towards various chemical inhibitors,

metallic ions and high concentration of sodium chloride (NaCl). It exhibited high Ki

value of 1000 mM with glucose. The kinetic parameters of Bgl1A displayed the Km of

0.39 mM and Vmax of 50.7 µmol mg-1 min-1.

Nam et al. (2010) demonstrated the cloning and sequencing of β-glycosidase gene from a

hyperthermophilic bacterium Thermus thermophilus KNOUC202, and was overexpressed

in a host E. coli JM109 (DE3), the cloned enzyme was completely characterized after

purification. The β-glycosidase gene consisted of 1,296 bp nucleotides encoding an active

protein of 431 amino acid residues, the sequence homology analysis confirmed that

recombinant enzyme was a member of GH family 1. The purified enzyme was

monomeric with a MW of 49 kDa, and exhibited a single band on polyacrylamide gel

electrophoresis. Enzyme had broad range of substrate specificity, Km value of pNPG was

lower as compared for the pNPGal and oNPG, the catalytic efficiency (kcat/Km) for pNPG

hydrolysis higher than those for pNPGal and oPNPG, though kcat value of recombinant

enzyme for the hydrolysis of pNPG was highly less as compared for the pNPGal and

Page 61: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

33

oNPG. Enzyme revealed optimal activity at 90°C and at the pH of 5.4. The cloned

enzyme presented great stability at 80-100°C temperature, and displayed 100% catalytic

activity after being incubation at 80°C for 120 min in pH 6.8 buffer.

Chang et al. (2011) isolated a bacterium from shrimp shell due to having great β-

glucosidase activity. The gram-negative bacterium belonged to Exiguobacterium genus,

named Exiguobacterium sp. DAU5. The gene comprised 1,350 bp encoded 450 amino

acids, was isolated from the genomic DNA of Exiguobacterium sp. DAU5 and

successfully cloned and expressed in host cells. SDS-PAGE indicated that the purified

β-glucosidase (BglA) enzyme had MW of 52 kDa. The alignment of amino acid

residues sequence homology with other bacterial β-glucosidases showed that enzyme

was a member of family 1. The BglA enzyme revealed that the best substrate for

catalytic activity was pNPG, at optima of temperature 45°C and optima of pH 7.0.

Hydrolytic efficiency was strongly influenced by metallic ions such as Zn2+, Ni2+, K+,

Na2+, Ca2+, Li+ and Mg2+. Chelating agent EDTA partially repressed the catalytic

efficiency of BglA.

Kim et al. (2011) reported a study on cloning of a β-glucosidase gene isolated from

Dictyoglomus turgidum and transformed E. coli strain of ER2566 with recombinant β-

glucosidase gene-pET28a(+) plasmid. This β-glucosidase belonged to GH3 that was

confirmed after the sequence analysis. The enzyme was purified using His-Trap affinity

chromatography, with a specific activity of 31 U mg-1. Purified monomeric enzyme had a

MW of 86 kDa, and showed peak activity at pH 5.0 and 85°C. Enzyme exhibited high

thermostability with a half-life of 334 min at 85°C. Enzyme had a broad substrate

specificity towards aryl-glycoside substrates with maximum hydrolytic activity with

pNPG with a Km value of 1.3 mM and kcat value of 13900-1 and also showed affinity with

oNPG, pNPGal, pNPX and pNPF. The effect of additives revealed that enzyme was

completely independent of metallic monovalent or divalent ions and EDTA.

Xu et al. (2011) cloned, sequenced and expressed a new heat stable β-glucosidase (bgl)

from a thermophilic filamentous fungus Chaetomium thermophilum, bgl gene contained

2,604 bp open reading frame (ORF) which encoded 867 amino acids having a potential

Page 62: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

34

signal of secretion. Recombinant β-glucosidase (Bgl) was achieved by expression of

cloned gene in Pichia pastoris. Recombinant enzyme was purified to homogeneity with a

MW of 119 kDa by SDS-PAGE analysis. Enzyme showed optimal catalytic activity at

pH 5.0 and 60°C, exhibited a half-life of 55 minutes at 65°C, and was quite heat stable up

to 50–60°C, retained 67.7% and 29.7% remaining activity after being incubated at 60°C

for 60 min and at 70°C for 10 min, respectively.

Haq et al. (2012) cloned a highly thermostable β-glucosidase gene of 1,341 bp from a

heterotrophic, obligate anaerobe Thermotoga petrophila RKU-1 (TpBglA). Heterologous

protein expressed in E. coli strain BL21 CodonPlus, purified monomeric recombinant

TpBglA enzyme displayed a prominent band of 51.5 kDa. The calculated Vmax and Km

values of TpBglA towards pNPG substrate were 42,720 µmol min-1 mg-1 and 2.84 mM,

respectively. Enzyme was optimally active at 80–90°C with a high specific activity of

30,400 U mg-1 against pNPG. TpBglA exhibited a broad substrate specificity towards

other p-nitrophenyl substrates and cellobiose. Conceivable catalytic regions of TpBglA

against nitrophenyl substrates and cellobiose were proposed on the basis of docking

studies.

Jabbour et al. (2012) identified, cloned, sequenced and characterized a thermo- and

glucose tolerant β-glucosidase enzyme from a heat loving bacterium

Fervidobacterium islandicum (FiBgl1A). The bgl1A gene contained 1,380 bp encoding

an enzyme of 459 residues and exhibited high homology to the other proteins of GH

family 1. The gene was expressed in E. coli host after ligated in pET-Blue1 vector, a high

level of FiBgl1A heterologous protein expression was observed, crude cell extract was

purified by heat treatment and gel filtration chromatography. Purified FiBgl1A with a

MW of 50 kDa, displayed optimal catalytic activity towards pNPG at 90°C and at pH

6.0–7.0. FiBgl1A showed remarkable thermostability over a broad range of temperature

(80-100°C). Purified enzyme exposed significant tolerance to glucose with Ki value of

211 mM, no metallic cations were required for the catalytic efficiencies, and not

influenced in the presence of EDTA, dithiothreitol, p-chloromercuribenzoate and urea

with 10 mM concentration. By the addition of various solvents (benzene, n-hexane,

hexadecane, heptane, toluene and alcohol) and surfactants except SDS, FiBgl1A activity

Page 63: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

35

was enhanced. Enzyme did not display any influence in the presence of different

saccharides especially 50 mM xylose.

Kim et al. (2012) identified and cloned a gene of β-glucosidase (1,467 bp) encoding 489

amino acid residues from Sulfolobus solfataricus, followed by overexpressed in E. coli

ER2566. The enzyme was purified by applying crude sample to a His-Trap High

Performance chromatography, finally the purity of enzyme analyzed on SDS-PAGE with

a band of 57 kDa. The purified enzyme displayed maximum hydrolytic activity at 90°C

and citrate phosphate buffer pH 5.5 with highest specific activity of 179 U mg-1 for

daidzin. Enzyme was highly stable at high temperature and exhibited at 95°C a half-live

of 10 h. With all these features, this cloned β-glucosidase from S. solfataricus was very

useful industrial biocatalyst among other β-glucosidases which can hydrolyzed isoflavone

glycosides.

Liu et al. (2012) cloned an internal sequence of a thermo-tolerant native β-glucosidase

(bgl3) gene from Aspergillus fumigatus Z5, a decomposing fungal strain. The internal

gene sequence contained an ORF of 2,622 bp nucleotides, which encoded a functional

protein, the predicted molecular mass was 91.47 kDa, and deduced protein belonged to

the family 3 of glycoside hydrolase. Cloned β-glucosidase (Bgl3) was successfully

expressed in Pichia pastoris X33, and heterologous protein was purified using affinity

column chromatography. Bgl3 purity was analyzed by SDS-PAGE that showed a band of

130 kDa MW. Characterization of purified recombinant Bgl3 was similar to the native

Bgl3 enzyme, had optimal temperature 60°C and pH 6.0. The cloned Bgl3 was quite

stable at 50-70°C temperature and 4.0-7.0 pH. Recombinant Bgl3 exhibited maximum

hydrolytic activity towards a synthetic substrate pNPG, with 101.7 U mg-1 specific

activity. Activity of enzyme was enhanced in the presence of metal cations; however

EDTA, SDS and Triton X-100 did not cause any obvious influence on enzyme. Km value

of recombinant Bgl3 for pNPG (1.73 mM) and for cellobiose (2.20 mM) were obtained

which was similar with native enzyme while Vmax values acquired towards these

substrates under standard assay conditions were lower than the native enzyme.

Page 64: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

36

Pei et al. (2012) cloned a β-glucosidase from a thermophilic bacterium

Thermoanaerobacterium thermosaccharolyticum DSM 571. The β-glucosidase (bgl)

gene encoded a protein of 443-amino-acids which commendably overexpressed, purified

and characterized in host cells of E. coli. The recombinant BGL protein expression level

in LB medium was improved from 6.6 to 11.2 U mg-1 by substituting the rare codons of

the N-terminal. Protein was purified by two-step methods (thermal precipitation and

affinity chromatography), and purified BGLA had 52 kDa MW. Optimal pH and

temperature of enzyme was 6.4 and 70°C, respectively. At 68°C, enzyme displayed a

half-life of 60 minutes and pH stability over a range of 5.2-7.6, Mn2+ and Fe2+ cation

significantly enhanced the BGLA activity. The Km of 0.62 mM and 7.9 mM were

observed for pNPG (Vmax of 64 U mg-1) and cellobiose (Vmax of 120 U mg-1), respectively.

Recombinant enzyme exhibited great tolerance to cellobiose and glucose; BGLA activity

was activated lower than 200 mM glucose concentration, while increasing concentration

of glucose, the catalytic activity was gradually repressed with Ki value of 600 mM

glucose.

Zou et al. (2012) reported the cloning and overexpression of a β-glucosidase (DtGH)

from Dictyoglomus thermophilum in E. coli BL21 using pET-21a vector. Induced the

recombinant culture with low concentration of IPTG followed by kept the culture at 37°C

for gene expression. Crude enzyme was purified by heat precipitation and His-Trap

affinity chromatography, and the finally purified fraction exhibited a single band (51.8

kDa) on SDS-PAGE. Purified DtGH biochemical characterization revealed that enzyme

had peak activity at 90°C temperature and neutral pH. DtGH showed great thermal

tolerance at 70-90°C with a high half-lives of 5 h at 90°C. The kinetic parameters of

DtGH towards pNPG were determined, Km and kcat value of 1.15 mM and 218 s-1,

respectively. Due to high thermostability, it was used for direct n-octanol glycosylation at

70°C and increased glucose conversion by 27%. Enzyme showed tremendous stability in

glucose and octanol-aqueous system, and retained 70% of its original activity even after

incubation at 70°C for 7 days.

Bai et al. (2013) reported molecular cloning of a β-glucosidase gene from thermophilic

bacterium Caldicellulosiruptor bescii (CbBgl1A). Purification of recombinant enzyme

Page 65: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

37

was done using heat treatment and chromatographic techniques, purified CbBgl1A

exhibited peak activity at 85°C and pH 6.8. CbBgl1A belonged to GH family 1, could

hydrolyzed a broad range of substrates such as aryl-β-glycosides and

cellooligosaccharides, and showed maximum affinity towards 4-nitrophenyl β-D-

glucopyranoside (pNPG) with 3.17 mM value of Km and 84.0 s−1 mM−1 value of kcat/Km.

The kinetic assay, molecular modeling and substrate docking of CbBgl1A with pNPG

and cellobiose which elucidated the weak binding ability of CbBgl1A with cellobiose.

Recombinant enzyme had high Ki value (113.8 mM) which revealed its ability to tolerate

the high glucose concentration.

Karnaouri et al. (2013) has been described an attractive report on cloning of a glycosyl

hydrolase (GH) family 3 β-glucosidase gene (bgl3a) from Myceliophthora thermophila.

The bgl3a gene was cloned under the control of a strong promoter and expressed in

Pichia pastoris. The purified fraction displayed a noticeable band of 90 kDa on SDS-

PAGE. The soluble active recombinant protein exhibited specific activity of 41 U mL-1

after 8 days induction. The purified MtBgl3a enzyme had the ability to catalyzed low

MW polysaccharides and substrates. MtBgl3a optimally hydrolyzed p-β-NPG at 70°C

optimal temperature and pH 5.0. Purified enzyme had displayed 143 minutes of half-life

at 60°C. Km value was 0.39 mM and 2.64 mM with p-β-NPG and cellobiose, respectively.

MtBgl3a showed great tolerance against D-gluconic acid, D-glucose and D-xylose sugars,

enzyme exhibited transglycosylation activity in the presence of methanol and its catalytic

activity was increased by short chain alcohol. Enzyme stability in ethanol was highly

prominent, and after 6 h incubation MtBgl3a retained 50% remaining activity as

compared to its original.

Li et al. (2013) described gene cloning of a thermophilic β-glucosidase from an archaeon

Thermofilum pendens (Tpbgl). The Tpbgl gene was overexpressed in an excellent host E.

coli. The purified enzyme had 77.8 kDa MW and exhibited peak catalytic activity with

pNPG substrate at 90°C in pH 3.5 buffer. After 60 minutes incubation at 95°C only 50%

catalytic activity was remained and Tpbgl enzyme showed high thermotolerant behavior

in an acidic conditions (pH 3.5). This extremophilic β-glucosidase included in glucoside

hydrolase family 3, the kinetic studies of recombinant Tpbgl revealed that enzyme had

Page 66: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

38

optimal catalytic efficiency (kcat/Km) of 3.05 with pNPG, Km value of 0.149 and 0.147

against cellobiose and mannobiose, respectively.

Lu et al. (2013) isolated a gene of β-glucosidase unbgl1A from a metagenomic library.

After amplification, unbgl1A gene was cloned and expressed in E. coli BL21. The

soluble cloned enzyme was purified by one-step method using affinity chromatography, a

band of 52 kDa MW was identified on SDS-PAGE followed by its biochemically

characterization was done. Under optimal temperature 50°C and pH 6.0, the enzyme

unBgl1A had a Km of 2.09 mM, and Vmax of 183.90 μmol min-1 mg-1. UnBgl1A activity

stimulated by NaCl, and had a great stability even at 600 mM of NaCl. The activity was

also enhanced by a variety of monosaccharides and disaccharides, can be tolerate up to

2000 mM glucose concentration, with a Ki value of 1500 mM.

Zhao et al. (2013) cloned a hyperthermostable β-glucosidase gene from Thermotoga

thermarum (Tt-bgl) and overexpressed in E. coli host. Recombinant host cells in LB-

medium induced at 37°C without IPTG, by this the activity Tt-BGL was improved to

13 U mL-1. The crude Tt-BGL was purified by using heat precipitation and affinity

chromatography. The purified enzyme had optimal activity at 90°C temperature and at

pH 4.8, and had a single band of 55 kDa on SDS-PAGE. The Tt-BGL showed high pH

and thermal stability. The Vmax and Km for p-nitrophenyl-β-D-glucopyranoside was 142 U

mg-1 and 0.59 mM, respectively. The enzyme was activated by glucose at ˃400 mM

concentrations, the Tt-BGL activity was progressively inhibited with further increasing

the glucose, but remained 50% of relative activity even in 1500 mM glucose.

Fang et al. (2014) demonstrated gene cloning and characterization of a β-glucosidase

(bglX) from Lactococcus sp. FSJ4. The bglX gene consisted of 1,437 nucleotides which

encoding 478 amino acid residues, purified recombinant monomeric bglX showed a

single band of 54 kDa by SDS-PAGE analysis. The bglX displayed broad substrate

specificity towards pNPG, pNPGal and pNPX which revealed that bglX was a

multifunctional biocatalyst. The enzyme showed great affinity towards aryl glycosides of

glucose and xylose. Optimal temperature and pH of enzyme was 40°C and 6.0 with

pNPG. Substrate docking and molecular modeling showed its multifunctional behavior

Page 67: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

39

against pNPG, pNPGal and pNPX. The active site pocket revealed the structure-function

relationship of bglX. Docking results elucidated that Glu 180 and Glu 377 were the vital

catalytic amino acids residues of recombinant enzyme, the value of

CDOCKER_ENERGY indicated that bglX has maximum activity towards pNPX

followed by pNPG and pNPGal. The experimental investigation are in conformity with

the obtained docking results.

Mehmood et al. (2014) demonstrated the gene cloning of a β-glucosidase (bglA)

belonging to glycoside hydrolase family 1 (GH1) from Thermotoga maritima and

overexpression in E. coli M15 host strain. Purified heterologous β-glucosidase (BglA)

displayed a prominent single band of 51 kDa on SDS-PAGE. The expression of

recombinant protein was enhanced when lactose used as an inducer, the soluble BglA

produced 40-47% of total cellular proteins which was 440 mg L-1 and 130 mg L-1

recombinant enzyme in Dubos salt medium and LB medium, respectively. The peak

activity of BglA was observed between temperature and pH range of 80 to 100°C and 5.0

to 7.0, respectively. The enzyme exhibited high thermostability up to 140°C. BglA had

Km, kcat and Vmax values of 0.56 mM, 187.1±20 s-1 and 238±2.4 IU mg-1, respectively.

BglA showed a half-life at 80, 90 and 100°C for 136 h, 71 h and 12.6 h, respectively.

Thermodynamic studies of BglA explained the enthalpy (33.73 kJ mole-1), entropy (-

246.46 kJ mole-1), melting temperature (130°C), Gibb’s free energy (127.96 kJ mole-1),

and activation energy for inactivation (36.92 kJ mole-1).

Schroder et al. (2014) characterized a thermotolerant archaeal β-glucosidase (Bgl1)

enzyme belongs to GH family 1 which was isolated from a hydrothermal spring

metagenome. Bgl1 displayed broad substrate specificity with high affinity towards

cellotriose, cellobiose and lactose. Bgl1 showed maximum specific activity 3195 Umg-1

at 90°C and pH 6.5. Enzyme was quite stable for 48 h at pH 4.5-9.5 and at 4°C. Bgl1

exhibited high thermostability, more than 40% activity was observed at 105°C, and after

30 min at 90°C a thermal activation was found. Thermal stability was 5 and 7-fold

enhanced after applying 100 and 200 bar pressure for 120 min at 90°C, respectively.

Enzyme activity was significantly increased by the addition of AlCl3, and Bgl1 also

Page 68: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

40

showed stability against glucose with Ki value of 150 mM. All these characteristics make

Bgl1 a prominent candidate for numerous applications at high temperature.

In 2014, Tang and co-workers cloned a β-glucosidase gene (bgl) from

Aspergillus oryzae GIF-10, effectively sequenced and overexpressed in Pichia pastoris.

The cDNA gene sequence consisted of 2,586 bp, encoding a protein of 862 amino acids

with a signal peptide chain. The high yield of protein (321 mg mL-1) was obtained after

seven days induction, and after purification recombinant β-glucosidase exhibited MW of

110 kDa on SDS-PAGE with the specific activity of 215 U mg-1. It exhibited broad

substrate specificity, efficiently hydrolyzed 1-4-α-diglycosides and 1-4-β-diglycosides.

Enzyme had 50°C optimal temperature, it displayed 65% remaining catalytic activity

after being incubated for 0.5 h at 60°C. Optimal pH of enzyme was 5.0 and stable over a

broad range of pH from 4.0-6.5, in the presence of Mn2+ cation hydrolytic activity of

enzyme was enhanced.

Yang et al. (2014) has been reported the study of a thermostable β-glucosidase gene of

GH family 3 from Neosartorya fischeri P1 (NfBGL1), and successfully cloned,

overexpressed and characterized in an expression host Pichia pastoris in a 3.7 L

fermentor at high cell density. After purification, SDS-PAGE exhibited a recombinant

NfBGL1 of approximately 80 kDa, enzyme showed maximum specific activity of 2189 ±

1.7 U mg-1 at 80°C and pH 5.0 against pNPG as a substrate. Enzyme NfBGL1 was stable

at 70°C for 2 h, displayed great enzyme catalytic affinity towards xylan, cellobiose,

barley β-glucan, sophorose, salicin, amygdalin, laminiarin, glycitin, lichenin and p-

nitrophenyl substrates. NfBGL1 efficiently hydrolyzed β-1,2 glycosidic bond and

exhibited great bioconversion for soybean isoflavone glycosides into free forms.

Dikshit R and Tallapragada P (2015) characterized a partially purified β-glucosidase from

Monascus sanguineus. Jack fruit seed was found to the best substrate to obtained

maximum yield of enzyme. At 60°C temperature and acidic pH, enzyme was optimally

active. Kinetic studies of enzyme were calculated by using different concentration of

pNPG, and found 0.89 mM of Km value. The enzyme activity was influence by metal

ions, 14.8% activity was inhibited by CaCl2 and 69.8% repressed by KCl. Enzyme

Page 69: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

41

efficiency was inhibited by 100 mM glucose concentration by which pNPG hydrolysis

did not take place. Molecular weight (116 kDa) of partially purified enzyme was

analyzed by SDS-PAGE.

Gumerov et al. (2015) cloned a highly thermo-tolerant multifunctional beta-glycosidase

(a putative β-galactosidase) gene Asac_1390 from a thermoacidophilic crenarchaeon

Acidilobus saccharovorans and was overexpressed in a mesophilic host (E. coli).

Asac_1390 gene encoded 490 amino acid residues and exhibited high sequence

homology with family 1 glycoside hydrolases from the different members of heat loving

archaea. Recombinant enzyme was purified to homogeneity with 55 kDa band, peak

catalytic activity was observed at (pH 6.0) 93°C. Asac_1390 enzyme showed extreme

thermostability, 50% activity was repressed at 90°C after 7 h incubation. Purified

Asac_1390 had a broad range of substrates specificity towards various aryl glycosides

and cellobiose, maximum hydrolytic activity was found for pNPGal (328 U mg−1) and

oNPGal (325 U mg−1) followed by pNPG (246 U mg−1) and pNPXyl (72 U mg−1).

Enzyme revealed great resistance to glucose, and Asac_1390 showed high Km value

(0.24 mM) and kcat/Km (1327 s−1 mM−1) for pNPG which demonstrated that Asac_1390

enzyme was not a β-galactosidase rather than a multifunctional β-glucosidase.

Ramani et al. (2015) identified a β-glucosidase isoform (Bgl4) from Penicillium

funiculosum NCL1, bgl4 gene encoding 857 amino acids which had a glycoside

hydrolase family 3 (GH3) catalytic module; successfully overexpressed in Pichia pastoris

KM71H, cloned Bgl4 protein was a glycoprotein which completely purified with a MW

of ~130 kDa. It had maximum activity at 60°C and pH 5.0 with 1354.3 U mg-1 specific

activity, and remain stable for 1 h at 60°C. Bgl4 displayed high activity towards aryl

substrates with β-xylosidic, β-glucosidic linkages and moderately hydrolyzed salicin and

cellobiose substrates. The enzyme exhibited maximum substrate conversion of p-

nitrophenyl-β-glucoside and cellobiose with the value of 3,332 and 2,083 μmol min-1 mg-

1, respectively. Bgl4 has exposed glucose tolerance up to 400 mM, and increased 2-fold

glucose yield when added with Trichoderma reesei Rut-C30 crude cellulase for cellulose

hydrolysis. Bgl4 was a thermostable and glucose tolerant enzyme, which could efficiently

use for bioethanol production.

Page 70: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

42

Xie et al. (2015) overexpressed a recombinant thermotolerant β-glucosidase gene in E.

coli from Thermotoga petrophila (TpBgl). By SDS-PAGE, it was confirmed that

recombinant purified TpBgl had MW of approximately 81 kDa. It was highly active at pH

5.0 and 90°C with the units of 21 U mL-1. Thermostability was enhanced in the presence

of Ca2+ cation. Enzyme had great selectivity for hydrolyzing at carbon C-20 of

ginsenoside Rb1, the inner and outer glucopyranosyl moieties, and produced ginsenoside

20(S)-Rg3 that was pharmacologically active. A highly thermotolerant β-glucosidase of

GH3-family from T. petrophila had improved thermostability by the addition of Ca2+.

Yang et al. (2015) reported a study on cloning of an intracellular β-glucosidase (BGL)

from Thermoanaerobacterium aotearoense, the gene (bgl) contained 1.314 kb, encoding

a 446 amino acid residual protein which belonged to the family 1 of glycoside hydrolase

(GH1) and had a canonical TIM barrel fold specific for GH1. To enhance the specific

activity of the crude enzyme, recombinant pET-bgl with chaperones propagated in E. coli

host which resulted in 9.2-fold increased expression (256.3 U mg-1) as compared to

without any chaperones. The purified enzyme showed highly thermo- and pH stability

with peak activity at pH 6.0 and 60°C. By the addition of 5 mM K+ or Na+, BGL activity

was significantly stimulated; enzyme exhibited great affinity towards cellobiose substrate

with the Km of 25.45 mM and Vmax of 740.5 U mg-1 values. In the presence of 50-250 mM

glucose concentration, BGL activity was stimulated and displayed the high Ki value of

800 mM glucose.

Zhao et al. (2015) has been cloned a novel GH family 3 β-glucosidase gene of 2.513 kb

from Myceliophthora thermophila (Mtbgl3b), which encoding 870 amino acid residues

and overexpressed in Pichia pastoris an efficient expression host. Highly active at 60°C

and pH 5.0, enzyme remained 90% activity at 50-65°C temperature and showed more

than 70% relative activity at acidic pH 3.5-6.0 and, MtBgl3b displayed psychrophilic

behavior and exhibited 51% relative activity at 40°C. MtBgl3b was quite stable at 60-

65°C, showed great affinity towards pNPG, and also had the ability to hydrolyzed pNPC,

cellobiose, cellotriose and cellotetraose. The Km 2.78 mM and Vmax 927.9 μM mg-1 min-1

kinetic values were determined. Finally, the enzyme catalytic reaction approach was

investigated by molecular docking strategy.

Page 71: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

43

Chan et al. (2016) identified a β-glucosidase gene (1359 bp) in the genomic DNA of

Anoxybacillus sp. DT3-1 (DT-Bgl), subsequently cloned and heterologously

overexpressed in E. coli BL21 (DE3). Phylogenetic study showed that DT-Bgl belonged

to GH family 1, the enzyme was effectively purified using Ni-NTA column

chromatography. DT-Bgl showed a single band of 53 kDa MW after analysis on SDS-

PAGE. Recombinant DT-Bgl enzyme was highly active, displayed peak activity against

pNPG and cello-oligosaccharides. The DT-Bgl exhibited best activity at slightly alkaline

pH 8.5 and 70°C, and did not need any co-factors or metallic ions for activation. The

values of Vmax and Km for DT-Bgl were 923.7 U mg-1 and 0.22 mM, respectively, using

pNPG as substrate. The DT-Bgl was a thermotolerant enzyme with low glucose

inhibition, and retained 93 % activity with the addition of 10 M glucose. It hydrolyzed

cellodextrins (long-chain) to cellobiose, and then converted cellobiose to glucose.

Long et al. (2016) cloned and expressed a 56,000 Daltons (Da) β-glucosidase (TthBgl)

from a hyperthermophilic bacterium Thermotoga thermarum, in a mesophilic host E. coli

BL21. Purified TthBgl showed maximum catalytic activity against pNPG (substrate) at

temperature 85°C and pH 5.0. After being incubated for 2 h at 80°C, enzyme retained

80% activity and displayed H+ ions stability from pH 5.0-6.0. TthBgl showed complete

independence of metallic ions while enzyme activity was considerably repressed by 1

mM Cu2+ and 0.1% sodium dodecyl sulphate (SDS), only 10% and 3% residual activity

was observed, respectively. The Km value was 2.41 mM and Vmax value of TthBgl was

8.79 U mg-1 against pNPG. By the addition of glucose, TthBgl activity was gradually

repressed, and 50% activity was reduced by 500 mM glucose. Phylogenetic analysis

confirmed that β-glucosidase belongs to GH family 3.

Peng et al. (2016) cloned and expressed a multifunctional gene belonging to GH family 1

from Caldicellulosiruptor owensensis (CoGH1A), which showed great activities of β-D-

glucosidase, β-D-galactosidase, β-D-xylosidase and exoglucanase. Enzyme exhibited

great thermostability at 75°C and stable for 12 h at this temperature. The enzyme

catalytic coefficients (kcat/Km) were 2467.5, 7450.0, 90.9, 1085.4, and 137.3 mM-1 s-1,

respectively against pNPG, pNPGal, pNPX, pNPC and cellobiose. CoGH1A was highly

active at pH 5.5 and 85 degree celsius with 1621 U mg-1 of specific activity. Km value of

Page 72: A THESIS TITLED - prr.hec.gov.pk

Review of Literature

44

CoGH1A towards pNPG, pNPGal, pNPC, pNPX and cellobiose were 1.52, 0.61, 0.87,

7.18 and 15.65 mM, respectively along with Vmax value of 4027, 5100, 1065, 736, 2424

μmol mg−1 min−1, respectively. Enzyme showed no apparent requirement for metallic

cation for its hydrolytic activity and remain active in the presence of 5-10 mM cations.

CoGH1A a multifunctional glycoside hydrolase family 1 enzyme has significant

competency in saccharification of lignocellulosic biomass and lactose decomposition.

Page 73: A THESIS TITLED - prr.hec.gov.pk

CHAPTER-III

MATERIAL & METHODS

Page 74: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

45

3.1. Materials

3.1.1. Chemicals, Enzymes and Kits

Mostly, all chemicals used in the present study were of high purity analytical grade

commercially available. Chemical reagents, detergents, inhibitors, salts and buffers were

acquired from Fluka (Switzerland) and Merck (Darmstadt, Germany), while all substrates

employed in the research work were purchased from Sigma chemicals Co. (St. Louis, MO,

USA). Antibiotic chloramphenicol, and cultivation media components were acquired from

Oxoid (Basingstoke, Hampshire, England). Enzymes used in molecular gene cloning, such

as T4 DNA ligase, restriction endonucleases, Polymerase chain reaction (PCR)

components along with Taq polymerase, and antibiotic ampicillin, X-gal, Isopropyl-β-D-

thiogalactopyranoside (IPTG), InsT/A cloning kit (Catalog # K1214), ready-to-use DNA

ladder (GeneRulerTM # SM0333) along with 6X DNA loading dye were acquired from

Thermo Fisher Scientific Inc. (Waltham, USA). QIAprep spin miniprep kit (Catalog #

27104) and QIAquick gel extraction kit (Catalog # 28704) were supplied by QIAGEN

(Hilden, Germany) to purify PCR amplicons and restricted DNA fragments from agarose

and impurities. Protein molecular size marker (Catalog # 69079-3) were obtained from

Novogen (Madison, WI, USA).

3.1.2. Bacterial Strains and Vectors

Hyperthermophilic bacterium Thermotoga naphthophila RKU-10T (ATCC BAA-489 /

DSM 13996 / JCM 10882), employed as a source of genomic DNA having β-1,4-

glucosidases encoding genes, was cultivated anaerobically. PCR amplicons of both TnbglA

and TnbglB genes were initially ligated in pTZ57R/T cloning vector (Thermo Fisher

Scientific Inc. Waltham, USA). Linearized ready-to-use pTZ57R/T vector tailed with 3'-

ddT over hangs, as Taq polymerase adds extra nucleotide adenine (A) 3'-ddA at the end of

amplicons during PCR, by this genes were easily cloned in vector. It was selected due to

one step cloning method, additional modifications of PCR products was not required, high

efficiency yields of recombinants (up to 90%), has multiple cloning site (MCS) designed

for easy manipulation of insert genes, and for effectual blue-white screening. Initially, E.

Page 75: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

46

coli DH5α strain was used as a host for recombinant plasmids propagation and

transformation. For over-expression, pET-21a(+) expression vector and mesophilic host E.

coli BL21 CodonPlus (DE3)-RIPL strain were used, which acquired from Novogen

(Madison, WI, USA).

3.1.3. General Growth Media

Usually, for the cultivating and maintaining E. coli DH5α and CodonPlus (DE3)-RIPL

bacterial strains LB medium was used supplemented with ampicillin (100 µg mL-1) when

required, cultivated at 37°C temperature with 200 revolution minute-1 (rev min-1)agitation.

LB-broth medium composed of 1% (w/v) NaCl, 0.5% (w/v) yeast extract and 1% (w/v)

tryptone, while LB-agar medium contained 1.5% (w/v) agar along with above mention

ingredients. The media components were mixed thoroughly in the double distilled water

(ddH2O) followed by autoclaved at temperature 121°C, pressure 15 lb in-2 for 20 minutes

in Erlenmeyer flasks.

3.1.4. Buffers and Reagents

In the present research work various buffers, reagents and solutions were used. All recipes

of the solutions and reagents are discussed in this section.

3.1.4.1. General stock solutions

Some general stock solutions were prepared in this study, which used to make various other

working solutions.

3.1.4.1.2. Tris–hydrochloride buffers (1 M, pH 7.6 and 8.0)

For the preparation of 1 M Tris-Cl (Tris-HCl), trizma base (molecular weight, 121.14 g

mol-1) dissolved in distilled water, adjusted the required pH (7.6 and 8.0) using 0.2 N HCl,

made the final volume by distilled water (dH2O), filtered the stock solutions.

3.1.4.1.3. Ethylenediaminetetraacetic acid (0.5 M, pH 8.0)

EDTA stock solution was prepared by dissolving EDTA powder salt (MW, 360.32 g mol-

1) in dH2O, slowly added 10 N NaOH in the solution and adjusted the required pH (8.0),

Page 76: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

47

when the solution become clear and pH adjusted, made the final volume by dH2O. The

stock solution was stored at room temperature after wet heat sterilization.

3.1.4.1.4. Potassium acetate (5 M, CH3CO2K)

Weight out required potassium acetate salt (MW, 98.14234 g mol-1), dissolved in some

dH2O initially, and then raised the final volume with dH2O.

3.1.4.2. Solutions for the extraction of genomic DNA

Genomic DNA from T. naphthophila culture was extracted by phenol-chloroform method,

originally demonstrated by Sambrook et al., (1989). For DNA isolation process, following

buffers and reagents were prepared according to the standard protocol.

3.1.4.2.1. TEN Buffer

The buffer was prepared by mixing 10 mM sodium chloride (NaCl), 10 mM Tris-HCl (pH

7.6) and 1 mM EDTA (pH 8.0) in the distilled water. After thoroughly mixing, autoclaved

the buffer at 15 lb in-2 pressure and 121°C temperature, and stored at 4°C.

3.1.4.2.2. SET Buffer

In SET buffer, EDTA (50 mM) and Tris-HCl (50 mM) having pH 8.0 and 7.6, respectively

dissolved in ddH2O and then added 20% sucrose, the final volume was made using ddH2O

and store at 4°C after sterilization.

3.1.4.2.3. Sodium chloride (5 M NaCl)

Weigh out required NaCl salt (MW, 58.44 g mol-1), dissolved in dH2O properly, made the

final volume, and stored at room temperature after autoclaving.

3.1.4.2.4. Sodium dodecyl sulfate (25% SDS)

Weigh out SDS detergent powder, mixed in dH2O carefully and store at room temperature.

3.1.4.2.5. Lysozyme

Always prepared freshly, 10 mg mL-1 solution of lysozyme was prepared in TEN buffer.

Page 77: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

48

3.1.4.3. Solutions for Agarose gel electrophoresis

3.1.4.3.1. Tris-EDTA (TE) buffer

TE buffer mostly used to store genomic DNA and plasmids. To prepare buffer having pH

8.0, dissolved EDTA (1 mM, pH 8.0) and Tris-Cl (10 mM, pH 8.0) in double distilled

water, mixed thoroughly and store 4°C after wet heat sterilization.

3.1.4.3.2. Tris acetate EDTA (50X) buffer

Dissolved trizma base (2 M), EDTA (50 mM, pH 8.0) and acetate (1 M Glacial acetic acid)

in ddH2O, this solution has approximately a pH of 8.6, therefore generally not adjusted.

Filtered the stock solution after that sterilized completely by autoclaving (121°C, 15 lb in-

2 for 15 minutes), and stored at room temperature. Working solution for gel electrophoresis

was 1X TAE, which was mostly prepared freshly.

3.1.4.3.3. DNA loading buffer (6X)

This dye has xylene cyanol FF (0.03% w/v), bromophenol blue (0.03%w/v), EDTA (60

mM), Tris-Cl (10 mM, pH 7.6) and glycerol (60% v/v). Mixed all the components in ddH2O

and stored at -20ºC for further used. Tracking dye is mixed with DNA and RNA samples

during electrophoresis.

3.1.4.3.4. Ethidium bromide

Stock of ethidium bromide (10 mg mL-1) was prepared in double distilled water (ddH2O)

or in TE buffer, with continue stirring to sure that ethidium bromide had completely

dissolved. After proper mixing, transfer the dye solution in a dark bottle or wrap the

container tube with aluminum foil.

3.1.4.4. Solution for Competent Cell Preparation

Bacterial cells were made competent using 50 mM Calcium chloride (CaCl2) chemical

method. After weighing the powder of calcium chloride (MW, 110.98 g mol-1), mixed in

dH2O thoroughly, filtered and stored at 4°C after sterilization.

Page 78: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

49

3.1.4.5. Solutions for plasmid DNA isolation

Plasmid DNA from the harvested bacterial cells was isolated by alkaline lysis method.

Three main solutions were used in this method.

3.1.4.5.1. Alkaline lysis solution-I

Mixed EDTA (10 mM, pH 8.0) and Tris-Cl (25 mM, pH 8.0) in ddH2O, adjusted the pH

of the solution (8.0), autoclaved for 15 minutes followed by the addition 50 mM glucose

using 0.22 µm membrane filter in aseptic conditions and stored at 4°C for used in research

work, when required.

3.1.4.5.2. Alkaline lysis solution-II

This solution must prepared freshly, mixed 1% SDS and 0.2 N NaOH from the stock

solutions in autoclaved dH2O by gentle vortexing. Solution-II has pH around 12.0,

therefore no need to adjust.

3.1.4.5.3. Alkaline lysis solution-III

Mixed potassium acetate (5 M, 60 mL) and glacial acetic acid (11.5 mL) in ddH2O distilled

water to make final volume 100 mL. Adjusted the pH 5.0 of the solution, stored at 4°C

after wet heat sterilization.

3.1.4.6. Buffers and reagents for SDS-PAGE

Following buffers and reagents used for making SDS-PAGE resolving and stacking gel.

3.1.4.6.1. Resolving buffer (pH 8.8)

Required amount of Trizma base was dissolved in ddH2O to prepared 1.5 M Tris-Cl,

adjusted pH to 8.8 with HCl (0.2 N), afterward make the final volume.

3.1.4.6.2. Stacking buffer (pH 6.8)

Weigh out Trizma base for the preparation of 0.5 M Tris-Cl, dissolved in ddH2O, pH was

adjusted to 6.8 with HCl (0.2 N). Sterilization was obtained by autoclaving the solution

and stored at 4°C.

Page 79: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

50

3.1.4.6.3. Ammonium per sulphate (10% APS)

APS (10 % w/v) mixed in ddH2O, freshly made at the time of gel preparation.

3.1.4.6.4. Acrylamide/Bis-acrylamide Solution (30%)

Acrylamide (29 g) and bis-acrylamide (1 g) mixed well in ddH2O, stored in a brown bottle

at 4°C.

3.1.4.6.5. Sodium dodecyl sulfate (10% SDS)

SDS (10%, w/v) powder dissolved in ddH2O, and stored at room temperature.

3.1.4.6.6. Gel loading buffer (5X, pH 6.8^)

Loading dye sample buffer (5X) was prepared by dissolving SDS (10%, w/v),

bromophenol blue dye (0.25%, w/v), glycerol (50%, v/v), Tris-Cl (0.25 M, pH 6.8) and β-

mercaptoethanol (5%, v/v) or DTT (0.5 M Dithiothreitol) in ddH2O. Dye pH was

approximately 6.8 after mixing all the components. After preparation, the dye dispensed

aliquots (1 mL) in sterilized tubes and stored at -40oC. β-mercaptoethanol was added just

before use.

3.1.4.6.7. Tank/ Running buffer (1X, ~pH 8.3)

Also known as Tris-glycine buffer, 1X buffer was prepared by dissolving 1.47% (w/v)

Glycine, 0.3% Trizma base and 0.1% (w/v) SDS in dH2O. Adjusting the pH of the buffer

to 8.3 with 0.2 N NaOH or HCl before making the final volume, then filtered the buffer

and stored at room temperature. Mostly, buffer has ~pH 8.3 so no need to adjust.

3.1.4.6.8. Staining Solution

Acetic acid (10%), methyl alcohol (40%) and coomassie Brilliant Blue R-250 (0.1%)

dissolved in ddH2O. Filtered the solution and stored in an amber or brown colored bottle

at room temperature.

3.1.4.6.9. Destaining solution

Solution was made by mixing 10% acetic acid and 40% methyl alcohol in dH2O, and stored

in a brown bottle at room temperature.

Page 80: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

51

3.1.4.7. Buffers and reagents for Native-PAGE

Mostly same chemical reagents and buffer used in Native-PAGE except that SDS is not

applied during the formation of separating and stacking gel mixtures. Native-PAGE

loading dye buffer and Running buffer recipe is changed from the SDS-PAGE.

3.1.4.7.1. Native-loading buffer (2X, pH 6.8^)

2X native loading dye sample buffer was prepared by dissolving bromophenol blue dye

(1%, w/v), glycerol (25%, v/v), Tris-Cl (62.5 mM, pH 6.8) in ddH2O. Dye pH was

approximately 6.8 after mixing all the components, aliquots (1 mL) in sterilized tubes and

stored at -40oC.

3.1.4.7.2. Tank/ Running buffer (1X, ~pH 8.3)

1X glycine buffer was prepared by dissolving 1.47% (w/v) Glycine and 0.3% Trizma base

in dH2O. Mostly, buffer has ~pH 8.3 so did not adjust but if not then adjusting the pH of

the buffer to 8.3 with 0.2 N NaOH or HCl before making the final volume, then filtered the

buffer and stored at room temperature.

3.1.4.8. Buffers and Reagents for enzymes analysis

After cloning, various buffers used for the analysis and characterization of recombinant

enzymes.

3.1.4.8.1 Sodium acetate buffer (3.7–5.6)

This buffer is also known as Acetate buffer, to make this buffer, stock solution of 0.2 M

sodium acetate (C2H3NaO2) and 0.2 M acetic acid (CH3COOH) were prepared first, having

molecular weight of 82.0343 g mol-1 and 60.05 g mol-1, respectively. To make, 100 mL of

desired pH buffer (4.0–5.5), both stock solutions were adding in different ratio (volume,

mL) and make a final volume of 100 mL.

3.1.4.8.2. McIlvaine buffer (2.2–8.0)

McIlvaine buffer, composed of disodium hydrogen phosphate (0.2 M Na2HPO4) and citric

acid (0.1 M C6H8O7), most commonly known as citrate buffer or citrate phosphate buffer.

Page 81: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

52

Initially, the stock solutions of 0.2 M Na2HPO4 (MW, 141.96 g mol-1) and 0.1 M citric acid

(MW, 192.124 g mol-1) were prepared. After that, citrate phosphate buffer of desired pH

(from 2.5–8.0) was made by mixing both stock solutions to attained a final volume of 100

mL.

3.1.4.8.3. Tris-Cl buffer (7.0–9.0)

Weigh out Tris base and prepared 0.2 M Tris-Cl stock solution which was used later on to

made the required pH and molarity buffer with 0.2 N HCl.

3.1.4.8.4. Sorensen’s phosphate buffer (5.8–8.0)

The required pH buffer was acquired by mixing sodium dihydrogen orthophosphate (0.2

M NaH2PO4, 119.98 g mol-1 MW) and disodium hydrogen phosphate (0.2 M Na2HPO4)

stock solutions in different volumetric ratio and get 100 mL final volume of the buffer.

3.1.4.8.5. HEPES buffer (6.8–8.2)

4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (C8H18N2O4S, 238.3012 g mol-1 MW),

50 mM buffer was prepared in dH2O, with a pH of 7.0 to 8.0, maintained the required pH

by 10 N NaOH prior to make the final (100 mM) volume.

3.1.4.8.6. MOP buffer (7.0–7.8)

MOP is the famous name of 3-(N-morpholino) propanesulfonic acid (C7H15NO4S, 209.26 g

mol−1 MW), 50 mM buffer was prepared in dH2O, adjusted the pH with 10 N NaOH.

3.1.4.8.7. CAPS buffer (9.0–11.0)

CAPS is the common name of 3-(cyclohexylamino)-1-propanasulfonic acid (C9H19NO3S,

221.32 g mol-1 MW), 50 mM buffer was made by dissolving the required weight of CAPS

in dH2O, desired pH of the buffer was obtained with 10 N HCl.

3.1.4.8.8. MES buffer (5.6–6.6)

MES is the name of 2-(N-morpholino) ethanesulfonic acid (C6H13NO4S, 195.2 g mol-1), 50

mM buffer was prepared in dH2O and maintained pH with 10 N NaOH.

Page 82: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

53

3.1.4.8.9. Bradford’s reagent

To prepare dye-binding reagent G-250 (0.1 g coomassie brilliant blue) dissolved in 95%

ethanol (50 mL) followed by added 85% ortho-phosphoric acid (100 mL), mixed all the

component well and raised the volume up to 1 L with ddH2O. This reagent was kept at low

temperature (4°C) after filtration (twice) in a brown bottle.

3.1.4.8.10. Bovine serum albumin (BSA) stock

To make standard curve of BSA, various dilution were prepared from stock solution of

BSA. Stock solution of BSA (1 mg mL-1) was prepared by mixing BSA powder (100 mg)

in dH2O and make the final volume 100 mL. Filter the solution before used.

3.1.4.8.11 Sodium carbonate (1 M Na2CO3)

Weigh out the required amount to prepare 1 M sodium carbonate (MW 105.98 g mol-1),

mixed well in dH2O, store at room temperature after filtration.

3.1.4.8.12. DNS reagent

3,5-Dinitrosalicylic acid (DNS) reagent, prepared by dissolving all these following

components which were DNS powder (10.6 g), NaOH (19.8 g), Rochelle salts (306 g

sodium potassium tartarate, KNaC4H4O6·4H2O), 8.3 g of sodium metabisulphate

(Na2S2O5), 7.6 mL of phenol (before used melted at 50°C) and 1416 mL dH2O. Mixed

thoroughly, stored at room temperature after filtration in brown or amber bottle to avoid

photooxidation.

3.1.4.8.13. Metallic ion solutions

Metallic salts of sulphate, chloride, nitrate and phosphate, 100 mM solutions were prepared

in buffer and stored at room temperature.

3.1.4.9. Buffers for anion exchange chromatography (Resource Q column)

Two buffers used for anion exchange chromatography, buffer A (binding buffer) and buffer

B (elution buffer) which were prepared as follows:

Page 83: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

54

3.1.4.9.1. Binding buffer (Buffer A)

Tris-Cl (50 mM, pH 8.0) was prepared in deionized water. The buffer was kept at 4°C after

filter sterilization (0.45 µm filter).

3.1.4.9.2. Elution buffer (Buffer B)

The elution buffer B was prepared by dissolving NaCl (1 M) in Tris-Cl buffer (50 mM, pH

8.0). Stored the buffer at 4°C after filtration with 0.45 µm filter.

3.1.4.10. Buffers for Hydrophobic interaction chromatography (Resource ISO

column)

3.1.4.10.1. Binding buffer (Buffer A)

Binding buffer was prepared by dissolving (NH4)2SO4 (1.5 M) in Tris-Cl (50 mM, pH 8.0)

buffer. The buffer was kept at 4°C after filter sterilization (0.45 µm filter).

3.1.4.10.2. Elution buffer (Buffer B)

Tris-Cl buffer (50 mM, pH 8.0) was prepared for elution and stored the buffer at 4°C after

filtration with 0.45 µm filter.

3.2. Cloning and Sequencing of TnbglA and TnbglB Genes

To clone the cellulolytic genes from a hyperthermophilic bacterium T. naphthophila RKU-

10T in mesophilic expression host, first of all two putative cellulolytic (β-1,4-glucosidase)

genes were selected after studying nucleotide and FASTA sequences using online NCBI

database server followed by primers were designed to amplified the specific genes.

Genomic DNA was isolated after the cultivation of T. naphthophila RKU-10T, which was

used as a template for the amplification of β-1,4-glucosidase TnbglA and TnbglB genes.

Multiple copies of specific genes were obtained using PCR strategy, after amplification

PCR products were initially cloned in PTZR/T57 and then analyzed by sequencing of

nucleotides followed by sub-cloning in pET-21a(+) expression vector and propagated in

the mesophilic expression host. All steps related to the cloning are described in the

following sections.

Page 84: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

55

3.2.1. Sequence of β-1,4-glucosidase Genes

The complete genomic sequence of T. naphthophila was available on NCBI server under

GenBank accession number CP001839.1 and this bacterium has circular DNA with

1809823 base pairs (bp). The two putative β-1,4-glucosidase genes (1,341 and 2,166 bp)

sequence Tnap_0602 and Tnap_0656 locus tags were retrieved from T. naphthophila and

designated as TnbglA and TnbglB. Used gene sequences as query to carried out nucleotide

BLAST from NCBI.

3.2.2. Analysis of Non-Cutter Endonucleases

For gene cloning of TnbglA and TnbglB in pTZR/T57 and pET expression vectors, all the

non-cutter (zero-cutter) restriction endonucleases for both genes were determined using

NEBcutter V2.0 BioLabsInc software (Vincze et al., 2003). The nucleotide sequences of

both genes were uploaded to the online server and a list was retrieved which had all the

non-cutting restriction enzymes. By this analysis, NdeI and HindIII non-cutter restriction

endonucleases were selected for the cloning purpose of both genes. Therefore, NdeI

restriction site was introduced at 5′ end of both forward primers.

3.2.3. Primers Designing

To amplify the coding regions of TnbglA (ADA66698.1) and TnbglB (ADA66752.1), two

sets of primers were designed using Vector NTI Advance TM 10.3 software (Invitrogen

Corporation, California) and OligoCalc software (Kibbe, 2007). NdeI restriction site

upstream (underlined) in forward primer but no restriction site was added in reverse primer

because HindIII restriction site already present in multiple cloning site (MCS) of cloning

vector pTZ57R/T. After primers designing, the order was placed to Macrogen (Korea) after

that lyophilized primers obtained, which were diluted to make stock of 100 µM, and in

polymerase chain reaction (PCR) used 10 pM concentration of each primer.

3.2.4. Cultivation of Thermotoga naphthophila

A hyperthermophilic anaerobic bacterium Thermotoga naphthophila RKU-10T strain was

kindly provided by Prof. Dr. Camella Nesbo (Dalhouse University, Canada), employed as

Page 85: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

56

a source of two β-1,4-glucosidase genes TnbglA and TnbglB. The bacterium was cultivated

in MMI medium under anaerobic conditions by the method of Haq et al. (2012). The MMI

medium contained KH2PO4 (0.45 g L-1), MgSO4·7H2O (1.4 g L-1), KCl (0.33 g L-1), NH4Cl

(0.25 g L-1), NaCl (20.0 g L-1), soluble starch (1.0 g L-1), yeast extract (0.06 %),

CaCl2.2H2O (0.14 g L-1), MgCl2. 6H2O (0.9 g L-1), trace mineral (1.0 mL) solution (Widdel

et al., 1983), 0.02% resazurin (0.5 mL), and for anaerobic conditioning 0.5 M Na2S (4.0

mL). After mixing all the components thoroughly and adjusted the pH 6.8. The 125-mL

vials were used for the cultivation of T. naphthophila, sealed them tightly with completely

sterile stoppers of butyl-rubber. The culture was incubated at 80°C for 166 hours in a dry

incubator having static condition.

3.2.5. Extraction of Genomic DNA

Genomic DNA of T. naphthophila was isolated by the phenol-chloroform method which

was used as a template for the amplification of genes (TnbglA and TnbglB). All buffers

and reagents for the extraction of DNA were prepared essentially as described by

Sambrook and Russell, (2001). Bacterial culture (10 mL) was harvested in a sterile tube at

6,000×g (at 4°C for 10 minutes), the cell pellet was washed twice with TEN buffer (100

µL) and gently suspended in the same buffer after that centrifuged again (6,000×g at 4°C

for 10 minutes). Discarded the supernatant, dried the pellet then suspended the pellet in

SET buffer (150 μL) followed by added freshly prepared lysozyme (50 μL from the stock

of 10 mg mL-1), mixed gently and subsequently incubated at 37°C for 30 minutes, bacterial

cell wall break by this step. Afterward, 25% SDS (50 μL) was used to completely lyse the

cells. At this time, TEN buffer (100 μL) and 5 M NaCl (50 μL) was added sequentially.

Freshly prepared phenol-chloroform (1:1) was applied next in this mixture, by which

proteins were precipitated out. Now, the working tube was gently inverted to homogenize

all the components of the mixture, followed by centrifuged (6,000×g) at 4°C for

10 minutes. The upper viscous phase was transferred in a fresh sterile tube with the help of

a micropipette, added equal volume of chloroform in the supernatant and mixed it properly,

all residual phenol was removed by this step. The mixture was subjected to centrifugation

at 6,000×g (5 minutes), transferred the resulted upper aqueous layer in another sterile tube.

To precipitate DNA, treated this with double volume of absolute ethanol (chilled), and

Page 86: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

57

incubated for 15-20 minutes at -20°C to improved precipitation. After incubation, DNA

threads had seen clearly which was spooled out easily and transferred in a fresh tube or

centrifuged (10,000×g) at 4°C for 10 minutes, in order to get a DNA pellet. Then washed

resultant pellet with 70% ethanol and allowed to dry the pellet at 37°C. Resuspended both

spooled out DNA and DNA pellet in TE buffer (100 μL), added Rnase (1 μL) to the sample

tube to remove the RNA from the isolated DNA. Incubated the sample tube at 37°C for 1

h. After incubation, analyzed the DNA by absorbance measurements and quantification

followed by stored DNA at -40°C for further use.

3.2.6. Isolated DNA Analysis

Isolated DNA was analyzed by electrophoresis therefore, 0.8% (w/v) agarose gel was

prepared by mixing agarose with 1X TAE buffer followed by heating the solution in a

microwave oven for 1-2 minutes until it boiled and became transparent or completely clear.

Then cooled in a water bath at about 60-65°C, after that calculated amount of ethidium

bromide (0.5 μg mL-1) was added and mixed well. Poured the solution into a sealed gel-

casting platform followed by placed a suitable gel comb (well-former for sample loading)

in its slot to make wells. Allowed the gel to solidify completely at room temperature for

about half an hour. After solidification, transferred the agarose gel to the running tank of

the electrophoresis containing buffer and removed the comb cautiously. The genomic DNA

sample (5 µL) was mixed with 6X loading dye and electrophoresed through gel along with

DNA ladder (Figure 3.1). In 1X TAE buffer, electrophoresis was accomplished at 70 volts

for 55-60 minutes. When blue dye came near the bottom of the gel, stopped running. DNA

was visualized and analyzed by illuminating the gel on UV-transilluminator followed by

its image captured and saved the record using gel documentation system with GeneSnap

software (SynGene). Exposed the gel for only short time under UV light, to evade DNA

damage.

Page 87: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

58

Figure 3.1: GeneRuler DNA Ladder Mix, ready-to-use #SM0333 (0.1µg µL-1)

3.2.7. Quantification of DNA

DNA quantification or concentration of isolated DNA was determined by electrophoresis

(intensity of DNA band compared with the known standard amount) as well as

spectrophotometrically measuring absorbance at λ260 using TE buffer as a blank, in a UV-

spectrophotometer. However sample purity was analyzed by a ratio of A260/A280

(Sambrook and Russell, 2001). DNA quantity was obtained by using the following

formula.

Amount of DNA mL-1 = 50 × Optical density (OD260) × dilution factor

However, 1 OD260 = 50 µg mL-1 of DNA

3.2.8. Amplification of TnbglA and TnbglB

Primers were used to amplify TnbglA and TnbglB gene fragments containing 1.341 kb and

2.166 kb nucleotides using polymerase chain reaction (PCR). 50 µL reaction mixture was

prepared in an individual sterile PCR tube (0.5 mL) by placing on ice for each gene

amplification, mixture contained all the necessary components of the PCR along with

Page 88: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

59

specific concentration of each was stated in table 3.1. All Components of PCR were mixed

thoroughly and consolidated in the bottom of tube by brief centrifugation, and then

transferred to the new sterilized individual PCR tubes and a negative control was also

prepared for PCR analysis, containing all components except DNA template.

Table 3.1: Composition of amplification reaction mixture.

Reaction components Required concentration/Quantity

dNTPs mixture 0.2 mM

10X Taq buffer 1X Taq buffer

Forward Primer 10 pM

Reverse Primer 10 pM

MgCl2 2.5 mM

Template genomic DNA 40 ng

Taq DNA polymerase 1.5 U

Denuclease water to made 50 µL volume

3.2.9. Thermal Cycling Conditions

Placed the reaction mixture tube in a thermocycler for the amplification of DNA fragments.

Closed the lid of thermocycler, set its temperature 105°C, both TnbglA and TnbglB genes

were amplified using optimal thermocycler conditions. Therefore, optimal PCR program

was set according to the fragment length of both genes, and hence the thermal cycling

conditions for TnbglA and TnbglB varied from one another. The PCR optimal conditions

for TnbglA and TnbglB are described in figure 3.2a and 3.2b. To produce 3'-dA overhangs

in amplicons, final extension phase was used which enhanced the efficiency of ligation.

3.2.10. Analysis of PCR Products

To examine the amplicons, mixed the PCR product with 6X DNA loading dye and run on

0.8% agarose gel along with DNA size marker. The amplified DNA bands of genes

(TnbglA and TnbglB), which appeared after gel electrophoresis were visualized under UV

light. Following analysis, the amplicons band corresponding to 1.341 and 2.166 kb were

excised with a sharp sterilized surgical blade (to avoid to get extra agarose gel) and placed

Page 89: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

60

separately into two sterile pre-weighted 1.5 mL tubes to purify DNA fragments from

agarose gel.

3.2.11. Purification of Amplicons

To purify PCR amplified products from the agarose gel, QIAquick gel extraction kit was

used according to the recommended protocol. To melt the gel slices containing amplified

DNA bands after adding QG buffer (supplied with kit, as a gel melting solution), briefly

incubated the tubes in a pre-heated water bath at 50°C for 10 minutes. During the

incubation period, the tube contents mixed regularly with the intervals of 2-3 minutes by

95°C 95°C

52°C

72°C

4°C 5 min 1 min

1 min

1 min 10 min

30 cycles

Initial

Denaturation Denaturation

Annealing

Elongation Final Extension

72°C

Figure 3.2 (a): PCR cycling conditions for TnbglA

95°C 95°C

54°C

72°C

4°C 5 min 1 min

1 min

2 min 10 min

30 cycles

Initial

Denaturation Denaturation

Annealing

Elongation Final Extension

72°C

Figure 3.2 (b): PCR cycling conditions for TnbglB

Page 90: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

61

vortexing until the gel contents was completely dissolved in this buffer (did same with both

PCR amplicons). QG buffer solubilized the agarose gel slices and provided a suitable pH

condition (≤ 7.5) and salt for subsequent DNA binding to the silica-membrane. For DNA

binding, poured the sample mixture (700 µL) onto the QIAquick spin column present in a

collection tube (2 mL) and centrifuged (10,000×g) for 60 seconds. During this step, DNA

fragments adsorbed to the silica-membrane whereas all contaminants passed away through

the spin column. After discarding the flow-through, placed the column back, QG buffer

(100 µL) was poured onto the column and centrifuged (10,000×g) again for 60 seconds.

Afterward, bound DNA was washed with the ethanol-containing PE buffer (750 µL), and

centrifuged again for 60 seconds at 10,000×g. Discarded the flow-through, to remove the

residual ethanol (traces of PF buffer) from QIAquick column, that may interfere with the

following enzymatic reactions, centrifugation step was repeated again for 60-120 seconds.

Shifted the column into a fresh sterile eppendrof to elute the adsorbed DNA. Finally,

provided elution buffer was applied to the center of the column, and allowed it to stand at

room temperature for 120-180 seconds. Consequently, the bound DNA was eluted with EB

buffer (50 µL) after centrifuged at 10,000×g for 60 seconds. By measuring absorbance at

λ260 spectrophotometrically, concentration of purified DNA was determined. Visualized

and analyzed by agarose gel electrophoresis as explained earlier. After that stored the gene

clean products at -40°C for further use in TA cloning purpose.

3.2.12. Cloning of TnbglA and TnbglB in pTZ57R/T Vector

Initially, the purified amplicons of both genes were cloned in TA cloning vector pTZ57R/T

by employing tailing technique (Figure 3.3). By using this technique, PCR amplicons with

3'-dA overhangs, were directly ligated into linearized pTZ57R/T vector containing 3’-dT

overhangs. According to the recommended protocol of InsT/A clone kit, ligation mixture

(20 µL) was prepared in a sterile eppendrof tube (1.5 mL) by adding amplified product (50

ng) with linearized pTZ57R/T (100 ng). To break up any concatamers, both vector and

insert DNA were heated for few seconds in a pre-heated water bath at 65°C and quickly

chilled on ice. After chilling, added the 10X ligation buffer (2 µL, to get final concentration

1X) followed by T4 DNA ligase (5 units) and nuclease-free water, for volume made up.

Control reaction was also incubated parallel to the ligation reaction, containing no insert.

Page 91: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

62

All reaction mixtures were incubated at 16°C for overnight (16–18 hours). When ligation

were done successfully, the recombinant vectors designated as pTZ57R/T–TnbglA and

pTZ57R/T–TnbglB, which were employed to transform competent cells of E. coli DH5α.

Figure 3.3: Restriction map of pTZ57R/T, the vector pre-cleaved with Eco321 restriction enzyme and treated

with terminal deoxynucleotidyl transferase to make tailed with 3'-ddT overhangs at both ends to avoid re-

circulation of pTZ57R/T during ligation.

3.2.13. Preparation of Competent Cells

Component cells of E. coli DH5α were prepared according to the chemical (calcium

chloride) method with some minor modification, originally described by Cohen et al.

(1972). A single well isolated colony of E. coli DH5α was inoculated into sterile LB-broth

medium (10 mL) from freshly streaked LB-agar culture plate followed by kept at 37°C for

16 hours (overnight) in shaker at 200 rev min-1. The sterile LB-broth medium (50 mL) in a

Erlenmeyer flask was seeded with 1% (v/v) overnight (16 hours) culture and incubated at

37°C in a shaking incubator (200 rev min-1) until its OD600nm reached at 0.4-0.5 (mid log

phase). Culture was transferred to a fresh 50 mL sterile falcon tube (pre-chilled) after that

kept on ice bath for 10-15 minutes, E. coli DH5α cells must be keep cold during competent

cells preparation. Then, harvested the culture cells in a pre-chilled (at 4°C) Beckmen

centrifuge at 4,000×g for 10 minutes. After centrifugation, medium supernatant was

discarded and resuspended the pellet in ice-cold 50 mM calcium chloride (CaCl2) solution

Page 92: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

63

(half of the original culture volume) by gentle mixing. The cells suspension was incubated

on ice for 45–50 minutes, afterward centrifuged again as described above, decanted off the

supernatant and resuspended the resultant pellet in ice-cold 50 mM CaCl2 (1.5–2 mL) by

gentle swirling and kept on ice for further half an hour before transformation. Finally, 100–

200 µL aliquot of competent cells was transferred directly in sterile pre-chilled eppendrof

tube (1.5 mL) for transformation, while the remaining cells were stored at -80°C after

dispensed into aliquot, as 30% glycerol stock.

3.2.14. Transformation of E. coli DH5α

Ligation products (pTZ57R/T–TnbglA and pTZ57R/T–TnbglB) were used to transform

competent cells of E. coli DH5α according to the standard protocol of heat shock method

(Sambrook and Russell, 2001). Chemically prepared competent cells (200 µL) aliquots

were briefly thawed followed by adding overnight ligated DNA sample (5-10 µL), mixed

gently by tapping or pipetting and successively incubated on ice for 30-40 minutes. A

control transformation reaction was run parallel to the ligation product, containing a

circular vector (plasmid) without insert mixed with competent cells, and similarly

incubated on ice as describe above. After that, heat shock at 42°C was given to all mixture

tubes for 1.5 minutes and instantaneously incubated for 3-5 minutes on ice. Poured and

mixed well the sterile pre-heated (37°C) LB-broth medium (800 µL) thereafter to

facilitated the recovery of E. coli DH5α cells in a comfortable growth environment from

transformation shock. Followed by incubated the mixture for 1-2 hours at 37°C with

constant shaking. This incubation prior to spreading mixture on agar plates, allowed the

bacterial cells to recover completely and start to express their genes for antibiotic

resistance. After incubation, transformants (100 µL) were spread uniformly on LB-agar

plates supplemented IPTG (0.1 mM), ampicillin (100 µg mL-1) and X-gal (80 µg mL-1).

Remaining transformed cell suspension was subjected to centrifugation for 60 seconds at

10,000×g, approximately 800 µL supernatant was decanted off, remaining 100 µL used to

resuspend the cell pellet followed by spread on LB-agar plates and kept these plates at 37°C

for 16-18 hours (overnight). Several colonies were appeared on agar plates, while positive

putative transformants were identified by blue/white screening and further confirmed by

double digestion analysis, colony PCR and DNA sequencing.

Page 93: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

64

3.2.15. Colony PCR

Colony PCR was performed separately to confirm the positive transformants harboring

recombinant pTZ57R/T–TnbglA and pTZ57R/T–TnbglB plasmids. Prior to colony PCR,

selected ten to sixteen well-isolated white colonies for both cloned genes (TnbglA and

TnbglB), then made short streak or duplically spotted on freshly prepared LB-ampicillin

agar plates (100 µg mL-1) , and kept these plates at 37°C for overnight till the colonies

became 1-2 mm in diameter. Colony suspension of each selected single colonies were

prepared by transferring the colonies aseptically to the sterile individual eppendrof tubes

(1.5 mL) containing sterile water (50 µL), dissolved each colony in their respective tube

and boiled the suspension for 3-5 minutes to lyse putative cloned bacterial cells and

denature the proteins. Subsequently centrifuged (10,000×g) the all tubes for 2 minutes to

collect the condensate, all denature proteins and membranes were sedimented in the bottom

of the tube (pellet). Transferred the upper layer (supernatant) 10 µL from each colony tube

to properly labeled sterile PCR tubes (0.5 mL) and amplification reactions for bglA and

bglB were set up by adding all the components with exactly same concentration as

described above (Table 3.1) except template DNA and using gene-specific primers. Colony

PCR of TnbglA and TnbglB was carried out by applying same conditions as described

earlier. After amplification, the PCR products of all colonies were electrophoresed through

0.8% (w/v) agarose gel to examine the result, presence or absence of insert gene. Positive

bacterial transformants containing recombinant pTZ57R/T–TnbglA and pTZ57R/T–

TnbglB plasmids were maintained in E. coli DH5α with ampicillin as a pressure of

selection, stored at -80°C as glycerol stocks after restriction analysis of recombinant

plasmids.

3.2.16. Mini-preparation of Plasmid DNA

Recombinant pTZ57R/T–TnbglA and pTZ57R/T–TnbglB plasmids were isolated from

cloned DH5α culture using alkaline lysis method (Sambrook and Russell, 2001). PCR

positive colonies were streaked on freshly prepared LB-ampicillin agar plates (100 µg mL-

1), which were further used for making glycerol stocks and recombinant plasmid isolation.

Sterile LB-broth (10 mL) containing ampicillin (100 µg mL-1) was inoculated with a well

Page 94: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

65

isolated bacterial colony harboring recombinant plasmid in an Erlenmeyer flask. Incubated

the culture flask at 37°C for 16 hour (overnight) in an incubator with vigorous shaking (200

rev min-1). The inoculation and extraction of recombinant plasmid DNA procedure was the

same for all transformants. The overnight recombinant cultures were harvested in sterile

eppendrof tubes (1.5 mL) by centrifugation (4,000×g) for 3-5 minutes at 4°C, discarded

the resultant supernatants and dried the pellets. The pelleted cells were resuspended in ice-

chilled alkaline lysis Solution I (100 µL) by vigorously vortexing. Then, added freshly-

prepared alkaline lysis Solution II (200 µL), mixed the contents by gently vortexing or

inverting the tubes for 3-4 times followed by placed all tubes on ice for 3-5 minutes.

Thereafter ice-chilled Solution III (150 µL) was poured into the tubes and again incubated

on ice for further 10 minutes, then at 4°C centrifuged (10,000×g) the tubes for 10-15

minutes. Pellets were discarded while the resultant supernatants were transferred to the

new sterile eppendrof tubes (1.5 mL) subsequently added equal volume of

phenol:chloroform solution (1:1) to all tubes, and then by vortexing shake well, and

centrifuged (10,000×g, 4°C) for 3-5 minutes. Upper plasmid containing aqueous layer from

all tubes were carefully shifted to fresh sterile tubes, to remove residual phenol (if present)

from the separated aqueous layer, chloroform (equal volume) was poured to each tube and

centrifuged (10,000×g, 4°C) again for 3-5 minutes. Upper layer from all tubes were taken

out in fresh sterile tubes, precipitated the plasmid DNA by adding ice-chilled double

volume of absolute ethanol, kept all tubes on ice for 5 minutes, pelleted by centrifugation

(10,000×g, 4°C) for 5-10 minutes. Washed the precipitated DNA pellets with 70% ethanol

(1 mL), discarded the supernatant cautiously and air dried the pellets. Dissolved the

plasmid pellets in TE buffer (50 µL) containing DNase-free RNase (1 µL) and kept all

these tubes for further 60 minutes at 37°C after that analyzed the plasmids DNA by gel

electrophoresis and stored at -40°C for further use.

3.2.17. Restriction Analysis of pTZ57R/T–TnbglA and pTZ57R/T–TnbglB

Isolated recombinant plasmids (pTZ57R/T–TnbglA and pTZ57R/T–TnbglB) were used to

confirm the presence of insert genes by single and double restriction analysis with specific

molecular scissors, NdeI and HindIII. For restriction analysis of both recombinant

plasmids, reaction (50 µL) was prepared in individual fresh sterile eppendrof tubes (1.5

Page 95: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

66

mL) by mixing plasmid DNA (~15 µg), 10X tango buffer (2X final concentration), NdeI

(~10 units), HindIII (~10 units) and nuclease-free water, to made final volume 50 µL.

Mixed all the restriction reaction components by vortexing and reaction tubes incubated

for overnight (16 hours) at 37°C. The resulting DNA restriction pattern of both reactions

were analyzed under UV-light after electrophoresed on 0.8% agarose gel along with DNA

size marker.

3.2.18. Preparation of Glycerol Stock

After the conformation of positive transformants, all bacterial recombinant colonies were

maintained in the form of glycerol stocks after culturing. To prepare the glycerol stocks,

overnight culture of positive clones were mixed with sterile 30% glycerol (w/v) in a sterile

screw caped cryo tubes, immediately stored at -80°C after mixing properly, for the long

period.

3.2.19. Sequencing of Cloned Genes

To get the plasmid DNA of ‘sequencing-grade’, the recombinant plasmids (pTZ57R/T–

TnbglA and pTZ57R/T–TnbglB) were extracted according to the recommended procedure

of the miniprep kit (Qiagen). The overnight recombinant culture was harvested, and

resuspended the pellet in PI buffer (250 µL) followed by the adding P2 buffer (250 µL),

then gently inverted the tube, poured N3 buffer (350 µL) and again inverted the tube 3–4

times. Centrifuged (10,000×g) for 5 minutes at room temperature, discarded the pellet and

loaded the supernatant to the QIAprep spin column after that centrifuged (10,000×g) again

for 60 seconds. Discarded the flow-through, washed the column with PE buffer (750 µL),

centrifuged (10,000×g) again for 60 seconds, and discarded the flow-through. To eliminate

residues of wash buffer, repeated the centrifugation (10,000×g) step again. To elute bound

DNA, placed the column in a clean sterile tube (1.5 mL), poured TE buffer (50 µL) on to

the column, and allowed to stand for 120 seconds followed by centrifuged (10,000×g) for

60 seconds. Quality and quantity (concentration) of eluted plasmid DNA was scrutinized

by λ260 measurements and electrophoresis. Recombinant plasmids were provided to

Macrogen, Korea for genes sequencing.

Page 96: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

67

3.2.20. Sequence Analysis and Protein Structure study

By utilizing Blast server at NCBI and ClustalW2 (Clustal omega), nucleotide and amino

acid sequences of both recombinant proteins were analyzed (aligned and compared).

Clustal omega online server was used to find the homology with others related protein

sequences and conserved regions. Theoretical molecular mass, isoelectric point (pI),

positively and negatively charged amino acid residues, aliphatic index and instability index

was calculated for amino acid sequence using ProtParam tool. The deduced protein

sequences of TnBglA and TnBglB were modeled for 3D structure utilizing SWISSMODEL

(Kiefer et al., 2009) and RaptorX sever (Källberg et al., 2012). PDBsum (Laskowski, 2009)

was used to study the secondary structure of TnbglA and TnbglB; and some tools of

ExPASy were also used to study the peptide information, protein structure elucidation

(swissProt) and functional information (Gasteiger et al., 2003).

3.3. Sub-cloning of TnbglA and TnbglB in Expression Vector

To get the adequate level of TnbglA and TnbglB expression, an excellent expression vector

was selected from vector series, amongst which T7 expression system using pET vectors

in E. coli BL21 CodonPlus (DE3)-RIPL prokaryotic host, has proved to be an excellent

over-expression system. An efficiently strong T7 lac promoter has been developed using

pET expression system to regulate the foreign gene of interest expression in E. coli BL21

CodonPlus (DE3). Therefore, pET-21a(+) proficient expression vector was employed to

get the high-level expression of both inserted genes (Figure 3.4). Firstly, constructed the

TnbglA and TnbglB expression plasmids followed by propagated into DH5α and for

expression, BL21 CodonPlus strain of E. coli was used as a mesophilic prokaryotic

expression host, which contains extra copies of the proL, leuW, ileY and argU tRNA

genes.

Page 97: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

68

Figure 3.4: Map of pET-21a(+) expression vector with cloning and expression sites. Vector contain a

pBR322 plasmid origin of replication (Ori), gene for ampicillin resistance (ampr), T7 lac promoter, which

contain a 25 bp long lac operator sequence. HindIII restriction site for the insertion of genes also shown.

3.3.1. Construction of Recombinant Expression Plasmids

E. coli DH5α competent cells was transformed with the purchased pET-21a(+) expression

vector followed by prepared glycerol stocks of E. coli DH5α containing pET-21a(+) vector

and stored at -80°C. For construction of recombinant expression plasmid, pTZ57R/T–

TnbglA and pTZ57R/T–TnbglB recombinant plasmids as well as pET-21a(+) vector were

isolated from the E. coli DH5α glycerol stocks by using alkaline lysis method (Sambrook

and Russell, 2001). Recombinant plasmids (pTZ57R/T–TnbglA and pTZ57R/T–TnbglB)

and expression vector pET-21a(+) were double cleaved with NdeI and HindIII restriction

Page 98: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

69

enzymes. Double digestion reactions (50 µL for each) were prepared in individual fresh

sterile eppendrof tubes (1.5 mL) by mixing isolated recombinant plasmid DNA along with

all components with same concentrations for proper reaction, as described earlier. Mixed

all the restriction reaction components by vortexing and incubated at 37°C for overnight

(16 hours). With pET-21a(+) vector, a corresponding double digestion was also carried

out. After incubation, the restricted plasmid reactions were mixed with DNA loading buffer

(6X) followed by electrophoresed on 0.8% agarose gel along with the DNA size marker,

after running the gel plasmid double restriction analysis was evaluated under UV-light.

The digested fragments (bands) of inserted genes (TnbglA and TnbglB) as well as pET-

21a(+) vector were excised from the 0.8% agarose gel, immediately transferred into the

individual sterile tubes (1.5 mL) and removed from gel using QIA quick gel extraction kit

(protocol described earlier). Purification of insert genes and linearized pET-21a(+),

restricted gene fragments (TnbglA and TnbglB) and pET-21a(+) were ligated by using T4

DNA ligase at the corresponding sites. The ligation reaction of bglA and pET was set up

in sterile tubes (1.5 mL) by adding double digested insert TnbglA (500 ng), 10X ligation

buffer (1X final concentration), double restricted pET-21a (100 ng), T4 DNA ligase (5

units) followed by make the final volume with nuclease-free water, while the ligation

reaction of TnbglB had all the components with same concentrations as described above

except the purified insert bglB whose final concentration was 400 ng in the reaction. A

control reaction was also prepared which did not had insert. Mixed all the ligation

component in respective tubes by vortexing and spun down by centrifugation at 4,000×g

for 0.5 minute after that incubated at 16°C for 16 hours (overnight).

3.3.2. Transformation of E. coli DH5α with Constructed Expression Plasmids

Competent cells were prepared chemically using CaCl2, and ligation mixtures of both genes

(TnbglA and TnbglB) were used to transformed E. coli DH5α competent cells individually

by heat shock method (previously described). After transformation, resultant bacterial

transformants cells were plated over uniformly on separate LB-agar plates containing

ampicillin (100 µg mL-1), and then kept at 37°C for 16-18 hours, several white colonies

appeared on agar plates. Positive putative transformants harboring recombinant plasmids

Page 99: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

70

(pET-21a–TnbglA and pET-21a–TnbglB) were screened and separated by colony

polymerase chain reaction and further confirmed by restriction analysis.

3.3.3. Colony PCR and Extraction of Recombinant Expression Plasmids

Putative transformants colonies were used to perform colony PCR separately for both

genes according to the protocol as described earlier. After colony PCR, positive bacterial

transformants harboring recombinant expression plasmids (pET-21a–TnbglA and pET-

21a–TnbglB) were analyzed and confirmed by electrophoresis. Successful clones were

streaked again and kept them on 4°C. Inoculated positive colonies of E. coli DH5α in LB-

broth containing ampicillin (100 µg mL-1) followed by kept in a shaking incubator for

overnight at 37°C, and extracted the recombinant plasmids by alkaline lysis method.

Visualized the quality and quantity of plasmids by agarose electrophoresis.

3.3.4. Restriction Analysis of Recombinant Plasmids

To further confirm the positive transformants, double digestion analysis of isolated

recombinant plasmids pET-21a–TnbglA and pET-21a–TnbglB were carried out in sterile

individual tubes (1.5 mL) using restriction enzymes NdeI and HindIII. Restriction reaction

was prepared separately by adding isolated recombinant plasmid (~15 µg), 10X tango

buffer (2X), NdeI (~10 units), HindIII (~10 units) and water (nuclease-free). Mixed and at

37°C kept the reaction mixture for 16 hours followed by analyzed on 0.8% agarose gel

electrophoresis. Positive clones were stored for long time as glycerol stock at -80°C, after

confirmation of positive recombinants by colony PCR and double restriction analysis.

3.3.5. Transformation of E. coli BL21 CodonPlus with Constructed Expression

Plasmids

Isolated recombinant plasmids (pET-21a–TnbglA and pET-21a–TnbglB) were used to

transform E. coli BL21 CodonPlus competent cells as an expression host by heat shock

method. After transformation, the bacterial transformed cells were plated on LB-agar plates

containing chloramphenicol (50 µg mL-1) and ampicillin (100 µg mL-1) followed by

incubated for overnight (at 37°C), a number of white colonies obtained on plates. To screen

positive transformed colonies, colony PCR was performed again at optimal standard

Page 100: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

71

conditions as described earlier. The positive transformants were stored at -80°C as stocks

for long time according to the same protocol as described earlier and used further for

production and expression analysis.

3.4. Heterologous Expression and Production of TnBglA and TnBglB

The expression analysis of both recombinant proteins were scrutinized by inoculating E.

coli BL21 CodonPlus harboring the pET-21a–TnbglA and pET-21a–TnbglB plasmids were

grown in sterilized LB broth and later on to enhance the production of both cloned proteins

various other modified media were also used, under the pressure of appropriate antibiotics

(ampicillin and chloramphenicol), and induced with isopropyl-β-D-thiogalatopyranoside

(IPTG) or later on with an alternative cheap inducer lactose was used for both recombinant

proteins production and over-expression.

3.4.1. Inoculum preparation

The seed cultures of both clones were prepared by inoculating individual LB sterilized

medium (10 mL) in two Erlenmeyer flasks (100 mL) with a 10 µL of frozen glycerol stocks

of engineered E. coli BL21 (DE3)-RIPL, supplemented with chloramphenicol (50 µg

mL−1) and ampicillin (100 µg mL−1) separately. Followed by both cultures were kept in a

shaking incubator at 200 rev min-1 for overnight (16 hours) at 37°C. These both seed

cultures were used as the standard inoculum to inoculate sterilized LB medium and all

other inducing media.

3.4.2. Production of TnBglA and TnBglB in LB medium

Initially, feasibility of LB-broth as the production medium for E. coli BL21 CodonPlus,

TnBglA and TnBglB expression and enzyme activity were investigated. Used 1% of

overnight both cloned cultures to refresh LB-broth in individual Erlenmeyer flasks

containing both antibiotic solutions with same concentration, kept at 37°C with 200 rev

min-1 till OD600nm reached at 0.6, the preferred phase for heterologous protein TnBglA and

TnBglB expression in our preliminary finding. Then heat shock was given to the bacterial

culture by placing all fermentation experimental flasks in a shaking water bath at 200 rev

min-1 at 42°C for 60 minutes. For TnBglA and TnBglB proteins over-expression,

Page 101: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

72

recombinant cells were induced with different concentration of inducer IPTG (0.1–1.0

mM) or lactose (10–250 mM), immediately after heat shock treatment. After induction, the

culture was allowed to incubate up to 72 hours, at various temperatures (16-42°C) in a

shaker (50-250 rev min-1); bacterial culture samples (fractions) were withdrawn after

regular intervals (2 hours) to assess the cell growth (OD600nm), optimal expression and

activity of heterologous protein TnBglA and TnBglB. By SDS-PAGE analysis,

heterologous proteins expression were scrutinized followed by assayed to determine

recombinant enzyme activity.

3.4.3. Enhanced expression and production using various inducing media

Small-scale expression analysis was set with various inducing media including LB+, ZB,

4×ZB, ZBM, ZYB, 3×ZYB, ZYBM9, 3×ZYBM9, and M9 with some modifications in

their composition were used in this present work (Studier, 2005). Individually, the effect

of ten various cultivation medium on recombinant bacterial growth, expression of

heterologous proteins (TnBglA and TnBglB) and activity were studied after inducing

individually either with IPTG or lactose. All used media with complete composition are

given in table 3.2. To enhance the expression and production of recombinant proteins or to

optimize the fermentation medium, inoculated all respective sterilized broth media (10 mL)

containing both antibiotics with the standard inoculum of recombinant E. coli BL21

CodonPlus (DE3)-RIPL, followed by overnight incubation in a shaker (200 rev min-1) at

37°C. Transferred 1% of all overnight cultures to their respective sterilized media

containing both antibiotics and incubated at 37°C (200 rev min-1), followed by done all

steps including heat treatment to sample fractions withdrawn, as described earlier. Here,

various parameters such as heat shock treatment, IPTG concentration, lactose

concentration, incubation temperature, agitation and time of incubation were studied to

enhance the activity and expression of heterologous proteins (TnBglA and TnBglB).

3.4.4. Analytical methods

Bacterial cell growth was monitored as the described by Basar et al. (2010). During

fermentation, culture broth sample were withdrawn regularly and at 4°C centrifuged

(12,000×g) for 10 minutes, obtained cell pellets were used to find out the cell growth.

Page 102: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

73

Resuspended the cell pellet in NaCl (0.9%, w/v), and OD600nm was measured when optical

density value exceeded 0.9, filtered the cell suspensions using membrane filter, then dried

the retentate in an oven (80°C) for 24 h, to calculate the dry cell weight (DCW).

Relationship of OD and DCW was assessed from many experiments, which showed that

one unit of optical density (OD600nm) was almost equivalent to 0.6 g DCW L-1.

Table 3.2: Compositions of different media.

Medium Composition

LB Yeast Extract (0.5%, w/v), NaCl (1.0%, w/v), Tryptone (1.0 %, w/v)

LB+ Yeast Extract (1.0%, w/v), NaCl (0.5%, w/v), Tryptone (1.6%, w/v).

ZB NaCl (0.5%, w/v), Tryptone (1.0%, w/v)

4×ZB NaCl (2.0%, w/v), Tryptone (4.0%, w/v).

ZBM NaCl (0.5%, w/v), Tryptone (4.0%, w/v)

ZYB Yeast Extract (0.5%, w/v), NaCl (0.5%, w/v), Tryptone (1.0%, w/v)

3×ZYB Yeast Extract (1.5%, w/v), NaCl (1.5%, w/v), Tryptone (3.0%, w/v)

ZYBM9

Yeast Extract (0.5%, w/v), NaCl (0.5%, w/v), Tryptone (1.0%, w/v),

NH4Cl (0.1%, w/v), KH2PO4 (0.3%, w/v), Na2HPO4 (0.6%, w/v),

MgSO4.7H2O (1 mM), and Glucose (0.4%, w/v).

3×ZYBM9

Yeast Extract (1.5%, w/v), NaCl (1.5%, w/v), Tryptone (3.0%, w/v),

NH4Cl (0.1%, w/v), KH2PO4 (0.3%, w/v), Na2HPO4 (0.6%, w/v),

MgSO4.7H2O (1 mM), and Glucose (0.4%, w/v).

M9

NH4Cl (0.1%, w/v), Na2HPO4 (0.6%, w/v), Glucose (0.4%, w/v), KH2PO4

(0.3%, w/v), NaCl (0.5%, w/v), MgSO4. 7H2O (1 mM), and CaCl2

(0.1mM).

3.4.5. Cell fractionation

Regularly, withdrawn bacterial samples were used to scrutinize the optimal expression,

activity assay and solubility of recombinant proteins (TnBglA and TnBglB) in all three

fractions: culture supernatant (extracellular), cell pellet (cell-bound) and cell lysate

Page 103: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

74

(intracellular). The recombinant cell culture (5 mL aliquot) collected by centrifugation

(12,000×g) at 4°C for 10 minutes and media supernatant was used as soluble (extracellular)

fraction. The pellet was washed twice and resuspended in Tris-Cl (50 mM) buffer of pH

8.0, disrupted cloned cells by keeping cell suspension on ice during sonication (10×30

seconds bursts with 60 seconds intervals between successive pulses) in a Ultra Sonicator

(UP 400s), then again centrifuged (12,000×g) at 4°C for 10 minutes and the cell lysate

(resultant supernatant) was used as soluble fraction (intracellular), and the pellet of cell

debris was resuspended in Tris-Cl (50 mM) buffer of pH 8.0, which constituted the cell-

bound fraction (insoluble). All soluble and insoluble fractions thus obtained, were

individually assayed to determine the maximum heterologous TnBglA and TnBglB protein

activity and expression.

3.4.6. Protein Estimation

Recombinant Soluble protein (TnBglA and TnBglB) concentration from all fractions were

assessed by either absorbing measurement at λ280 or dye-binding (Bradford) method in

which BSA used as a standard (Bradford, 1976). To estimate the protein in a given sample,

reaction mixture was prepared in well dry test tube by adding appropriately diluted protein

sample (1 mL) followed by Bradford reagent (5 mL), and control reaction had dH2O instead

of protein sample. In Bradford reagent, Coomassie brilliant blue G-250 (binding dye) bind

with basic amino acid residues of protein subsequently blue color develop. Incubated the

reactions at room temperature for 10 minutes prior to measure the absorbance at λ595 using

spectrophotometer after adjusting the control as a blank for auto-zero. A standard curve

was plotted with known concentration of a protein like BSA and applied to compute the

protein concentration in test sample.

3.4.6.1. Standard Curve of BSA

To make the standard curve of BSA for protein estimation, sequentially increased

concentration of BSA dilutions ranging from 02-100 µg were prepared, in properly labelled

individual test tubes by using stock solution of BSA (1 mg mL-1). In each dilution test tube,

Bradford reagent (5 mL) was added and shake well. After 10 minutes incubation at room

temperature, measured the absorbance of all dilution reactions (in duplicate) at λ595 using

Page 104: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

75

spectrophotometer against a blank reaction (adjusted to auto-zero) which contained

Bradford reagent and dH2O instead of BSA protein dilution. A graph was plotted by taking

absorbance values of all BSA dilutions at ordinate axis and concentration of BSA at

abscissa axis. Protein concentration from given sample was estimated from the slop of this

BSA standard curve (Figure 3.5).

Figure 3.5: Standard curve of Bovine Serum Albumin (BSA).

3.4.7. SDS-PAGE Analysis

The recombinant protein TnBglA and TnBglB expression and molecular weight (MW)

were determined by SDS-PAGE analysis, according to the described method of Laemmli

(1970). 12 % SDS gel was prepared in two phases, initially resolving gel mixture poured

into assemble casting plates followed by stacking gel mixture was poured.

3.4.7.1. Separating/Resolving gel preparation

To prepare 12% resolving gel (10 mL), all required chemical reagents and buffers as

described in table 3.3 except (TEMED) were mixed well in a fresh falcon tube (50 mL).

After mixing, TEMED was added mixed again by gently swirled and immediately poured

the resolving gel mixture between already assemble two plates of Bio-Rad mini-gel

electrophoresis vertical casting apparatus, at the top 0.5 inch (2 cm) space was left for the

stacking gel and poured dH2O (100 µL) at the top of gel to make a flat surface of the gel,

y = 0.0073x + 0.0153R² = 0.9996

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0 20 40 60 80 100 120

Ab

sorb

ance

(5

95

nm

)

Concentration µg mL-1

Standard Curve of BSA

Page 105: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

76

to prevent dehydration and to remove oxygen, which may inhibit polymerization. Kept the

gel along with caster undisturbed at room temperature for approximately 30 minutes for

gel polymerization followed by completely drained the upper layer of dH2O and prepared

stacking gel was loaded on resolving gel.

Table 3.3: 12% SDS-PAGE resolving gel composition.

Required Chemicals Volume (mL)

dH2O 3.3

1.5 M Tris-Cl (pH 8.8) 2.5

30% (w/v)Acrylamide/Bis-acrylamide 4.0

10% (w/v) SDS 0.1

10% (w/v) APS (freshly prepared) 0.1

TEMED 0.004

3.4.7.2. Stacking gel preparation

After polymerization of resolving gel, the second phase of SDS-gel called stacking gel (3

mL) was prepared by mixing all the chemical components listed in the following table 3.4

except TEMED. After properly mixing, TEMED was added, swirled and poured the gel

mixture at the top of gel assembly. Immediately insert the clean comb into the gel to make

proper wells for loading the samples, kept gel assembly undisturbed at room temperature

for 30 minutes to polymerize the gel.

Table 3.4: SDS-PAGE Stacking gel composition.

Required Chemicals Volume (mL)

dH2O 2.037

0.5 M Tris-Cl (pH 6.8) 0.380

30% (w/v) Acrylamide/Bis-acrylamide 0.5

10% (w/v) SDS 0.03

10% (w/v) APS (freshly prepared) 0.03

0.4% (w/v) Bromophenol blue dye 0.02

TEMED 0.003

Page 106: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

77

3.4.7.3. Sample preparation

Protein samples before subjected for SDS-PAGE were prepared by mixing protein sample

(10-15 µg) with 5X SDS-gel loading dye in sterile tube (1.5 mL) followed by boiling for

3-5 minutes in a boiling water (100°C) in order to denature sample proteins. Then,

centrifuged for 60 seconds and transferred the sample supernatant in fresh tube and loaded

in well.

3.4.7.4. Electrophoresis

After polymerization, the set gel cassette along with comb was taken out and clamped into

the electrophoresis tank apparatus. Poured Tris-glycine buffer (1X) in the tank and filled

up to the marked level of the tank subsequently the inner region of clamped apparatus was

also filled with the same buffer. Removed gel comb cautiously without disturbing the wells

followed by washed them with glycine buffer by using a syringe. The prepared samples

were loaded along with protein marker (Figure 3.6) into the individual wells carefully by

using a fine tip. Electrophoresis was performed at a constant current (2 mA cm-2) for about

90-120 minutes until from the bottom of the resolving gel bromophenol blue dye came out.

Figure 3.6: Novagen Perfect Protein Marker (10-225 kDa), Cat # 69079-3

Page 107: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

78

Turned off power supply immediately and removed the glass plates from the cassette and

separated out the gel carefully from the plates. To visualize the protein bands, stained the

gel with staining solution having Coomassive brilliant blue R-250, and placed the staining

gel tray in a shaker (50 rev min-1) for 30 minutes. After that placed out the gel from staining

solution, washed the gel with dH2O and dipped the gel in destaining solution with constant

agitation. Kept the gel on a shaking plate (50 rev min-1) for overnight to completely

destaining the gel, background became transparent and bands of protein were visible in the

gel.

3.4.8. Native-PAGE Analysis

The recombinant protein TnBglA and TnBglB were analyzed by Native-PAGE also. Native

gel was also prepared in two phases, initially resolving (separating) gel mixture poured into

assemble casting plates followed by stacking gel mixture was poured.

3.4.8.1. Separating gel preparation

To prepare 12% resolving gel (10 mL), all required chemical reagents and buffers as

described in table 3.5 except TEMED were mixed well in a fresh falcon tube (50 mL).

After mixing, TEMED was added mixed again by gently swirled and immediately poured

the resolving gel mixture between already assemble two plates of Bio-Rad mini-gel

electrophoresis apparatus. Kept the gel undisturbed at room temperature for

polymerization.

Table 3.5: 12% Native resolving/separating gel composition

Required Chemicals Volume (mL)

dH2O 3.396

1.5 M Tris-Cl (pH 8.8) 2.5

30% (w/v)Acrylamide/Bis-acrylamide 4.0

10% (w/v) APS (freshly prepared) 0.1

TEMED 0.004

Page 108: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

79

3.4.8.2. Stacking gel preparation

After polymerization of resolving gel, the second phase of native gel called stacking gel (3

mL) was prepared by mixing all the chemical components listed in the following table 3.6

except TEMED. After properly mixing, TEMED was mixed and poured the gel mixture at

the top of gel assembly. Immediately insert the clean comb into the gel to make proper

wells for loading the samples, kept gel assembly undisturbed for polymerization.

Table 3.6: Native stacking gel composition

Required Chemicals Volume (mL)

dH2O 2.1

0.5 M Tris-Cl (pH 6.8) 0.380

30% (w/v) Acrylamide/Bis-acrylamide 0.5

10% (w/v) APS (freshly prepared) 0.03

0.4% (w/v) Bromophenol blue dye 0.02

TEMED 0.003

3.4.8.4. Sample preparation

Protein samples before subjected for native PAGE were prepared by mixing protein sample

(~10 µg) with 2X native-gel loading dye in sterile tube (1.5 mL) and loaded in well.

Samples were not heated.

3.4.8.5. Electrophoresis

After polymerization, the set gel cassette along with comb was taken out and clamped into

the electrophoresis tank apparatus. Poured native Tris-glycine buffer (1X) in the tank and

filled up to the marked level of the tank subsequently the inner region of clamped apparatus

was also filled with the same buffer. Removed gel comb cautiously without disturbing the

wells followed by washed them with native glycine buffer by using a syringe. Loaded the

sample and run the gel with same method as described for SDS-PAGE.

Page 109: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

80

3.4.9. Determination of recombinant enzymes activity

The recombinant TnBglA and TnBglB enzyme activity assay were performed individually

in separate heat resistant caped tubes, using pNPG as a substrate and measured the amount

of pNP released from pNPG. A reaction mixture, contained 0.2 mL of pNPG (4 mg mL-1,

prepared in McIlvaine buffer of pH 7.0) and 0.4 mL of appropriately diluted enzyme (100

ng of BglA or BglB in 10 µL and 390 µL of pH 7.0 McIlvaine buffer) with 600 µL of final

volume. A single test was carried out in triplicate along with a control reaction, which had

all components with same concentration except recombinant enzyme (TnBglA or TnBglB),

under the assay conditions at 70°C for 10 minutes. All reactions were stopped by adding 3

mL of 1 M Na2CO3, followed by addition of diluted enzyme in control reaction tube and

to determine the released amount of pNP, measure the absorbance at 405 nm. A standard

curved of pNP was used to estimate the released amount of pNP and finally know about

the enzyme catalytic efficiency in term of units.

3.4.9.1 Standard Curve of para-Nitrophenol (pNP)

A standard curve of pNP was used to measure the enzyme activity of TnBglA and TnBglB.

This standard curve was drawn to convert the absorbance values into µM of pNP. Firstly,

prepared 1 mM p-nitrophenol of stock solution. Then, diluted the pNP serially ranging

from 20-500 µM in well dried label test tubes using stock solution, and made final volume

600 µL. A control was also run parallel by replacing pNP with dH2O, followed by adding

3 mL of 1 M Na2CO3 in each reaction tube and using spectrophotometer measured the

absorbance at 405 nm. A standard curve of pNP was plotted by taking absorbance along

Y-axis and various concentration of pNP along X-axis (Figure 3.7).

3.4.9.2. Enzyme Unit

“One unit of recombinant enzyme (TnBglA or TnBglB) activity corresponds to the amount

of enzyme necessary to release 1 μmoL (unit) of pNP per minute under the described

standard assay conditions”. Enzyme activity in term of units was calculated by applying

the following formula:

Page 110: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

81

Figure 3.7: Standard curve of para-nitrophenol (pNP)

3.4.9.3. Specific activity

Specific activity of TnBglA and TnBglB enzymes were described as enzyme units per mg

of the recombinant protein, and calculated by using the following formula:

3.5. Purification of Recombinant TnBglA and TnBglB

Various strategies were employed for the purification of recombinant β-1,4-glucosidases

enzyme after large scale (up to 3 L) production. After recombinant proteins production,

cultures were subjected to heat treatment, followed by ammonium sulphate NH4)2SO4

precipitation, dialysis and chromatography techniques were applied to purify cloned

TnBglA and TnBglB enzymes from the E. coli BL21 CodonPlus host proteins.

y = 0.0073x + 0.0153R² = 0.9996

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

1.8

0 50 100 150 200 250 300 350 400 450 500

Ab

sorb

an

ce (

40

5 n

m)

Concentration µM

Standard Curve of pNP

Conc. from graph (µM) × Dil. factor × 1000

Incubation time (min) × Total reaction volume (µL) Enzyme activity (U mL-1 min-1) =

U mL-1 min-1 of enzyme

mg of protein mL-1 Specific activity (U mg-1) =

Page 111: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

82

3.5.1. Batch production

Individually, the recombinant TnBglA and TnBglB enzymes produced on large scale at

optimal conditions which were found by small flask scale production. Initially, sterilized

LB broth medium was inoculated with 16 hours old overnight recombinant cultures

containing chloramphenicol and ampicillin with 50 µg mL-1 and 100 µg mL-1 as final

concentration, followed by left at 37°C in a shaking incubator for attaining the OD600nm of

culture 0.6. At 42°C, heat treatment was given for 60 minutes after that induced the cultures

with IPTG (0.5 mM) and kept flasks at 22°C with other optimal conditions for production

up to 72 hours. The bacterial culture was collected at optimal crude activity and expression

of recombinant TnBglA and TnBglB enzyme. Both enzymes were soluble, extracellular

and highly active, therefore crude supernatants were stockpiled after 72 hours and

concentrated twenty folds by using freeze drier or lyophilizer (Alpha 1-4 LO).

3.5.2. Heat Treatment

Lyophilized powder of both recombinant enzymes TnBglA and TnBglB from T.

naphthophila were resuspended in dH2O followed by subjected to thermal incubation

because both enzymes was isolated from hyperthermostable bacterium. Therefore, it was

observed that both enzymes must have thermal inactivation. To purify the recombinant

TnBglA and TnBglB, incubated the crude lyophilized enzymes in a pre-heated water bath

at 70°C for 60 minutes, after thermal precipitation the enriched enzyme samples were

immediately cooled at 4°C in an ice bath for 0.5 h, after that centrifuged (12,000×g) to

remove all denatured host heat labile proteins, for 20 minutes at 4°C.

3.5.3. Fractional Ammonium sulfate precipitation

Heat treated recombinant protein samples were subjected to ammonium sulfate

precipitation with 55–60% saturation according to the method described by De-Moraes et

al. (1999). Precipitation was performed at 4°C by slowly pouring the salt to the heat treated

partially purified enzyme samples with step wise increment and constant gentle stirring for

2-3 hours at 4°C, to attain various level of saturation. After each saturation stage,

centrifuged the samples at 12,000×g for 10 minutes at 4°C, to pellet down the precipitated

Page 112: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

83

recombinant proteins. Transferred the supernatant to another falcon and dissolved the pellet

in Tris-Cl buffer (50 mM) having pH 8.0, to ensure the precipitation of enzymes, activity

assay and protein contents were observed in each fractions. Stockpiled the fractional pellets

and dialyzed the resultant suspension to remove the salt from the precipitated protein.

3.5.4. Dialysis

Dialysis was carried out in order to remove the salt molecules from the precipitated protein

by using regeberated cellulose tubular membrane (Celluosep T4 #1430-33, membrane

filtration product Inc. USA) having nominal molecular weight cut off (MWCO) 12,000-

14,000. The dialyzing membrane was washed with buffer twice, and specific closure was

applied to close membrane tube one end. Followed by pouring the precipitated protein

suspension into tube with the help of micropipette, and also closed the other end of tube.

The dialyzing bag having sample suspended in pH 8.0 buffer of Tris-Cl (50 mM) of in a

beaker, placed the beaker on magnetic stir plate with continued stirring, regularly changed

the outer Tris–Cl buffer after 3–4 hours, process was continued at 4°C (approximately up

to 24 hours) till the suspension became clear and salt was completely removed. After

dialysis volume of the sample was increased, protein estimation and activity assay was

performed.

3.5.5. Anion Exchange Column (AEC) Chromatography

Followed by heat treatment, ammonium sulfate precipitation and dialysis to remove salt

both recombinant TnBglA and TnBglB were subjected to a Resource Q anion exchange

column (Amersham Pharmacia Biotech, Sweden) for anion exchange chromatography.

After studying the isoelectric point (pI) of cloned TnBglA and TnBglB enzymes using the

Protparam ExPASy tool, an appropriate choice was Tris-Cl buffer (50 mM, pH 8.0) as a

binding buffer (buffer A) and as an elution buffer or buffer B, 1 M NaCl salt mixed in 50

mM Tris-Cl buffer having pH 8.0 was prepared, both buffers kept separately after

sterilization at 4°C.

Initially, the column preparation was performed by washing Resource Q pre-packed

column with five column volume (5 CV) of double filtered (0.22 µm membrane millipore

Page 113: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

84

filter) deionized water at the rate of 1 mL min-1, followed by equilibrated with ten CV of

buffer A for approximately 10 minutes. The dialyzed sample was passed through a

millipore membrane filter (0.45 µm) before loading to avoid the blockage of column, and

the resulting enzyme fraction was applied to a Resource Q ion exchange column at a flow

rate of 3 mL min-1 and equilibrated with five column volumes (5V) of binding buffer (pH

8.0). At all steps, flow-through was collected, and during equilibration three unbound

fractions were collected. After washing, bound protein fraction was eluted from the column

with a 0-1 M NaCl linear increasing gradient at a flow rate of 1 mL min-1 and 0.6 psi

pressure. The elution profile was examined at 280 nm throughout the process. According

to elution peak, all collected fractions were analyzed separately by SDS-PAGE and activity

assay followed by active fractions pooled were together.

3.5.6. Hydrophobic Interaction Column (HIC) Chromatography

Resource ISO pre-packed column (Amersham Pharmacia Biotech, Sweden)

chromatography was selected for further purification. Resource ISO column was washed

with 3 column volume (3CV) of Tris-Cl 50 mM buffer of pH 8.0 containing 1 M (NH4)2SO4

(binding buffer A) and equilibrated with this buffer prior to applied active pooled fractions.

Before applying to the column, pooled active fractions solution was adjusted to 1.5 M

(NH4)2SO4 by adding salt, since the appropriate adsorption was occurred at high salt

concentration. Properly dissolved the salt in the sample followed by membrane filtration

down. After the application of sample (active fractions), the column was washed with the

same equilibration buffer till the UV absorption returned to near baseline at a flow rate of

1 mL min-1. The bound enzyme fraction was eluted with a 1.5-0 M (NH4)2SO4 decreasing

gradient. Flow rate was maintained at 2 mL min-1 during protein elution and measured the

absorption by online UV monitor. Across the major peak, various fractions were collected

and evaluated by SDS-PAGE analysis. Active enzyme was obtained as unbound fractions,

pooled together all active fractions of recombinant enzymes TnBglA and TnBglB. Purified

protein (TnBglA and TnBglB) concentration was determined by Bradford method and

thereafter purified recombinant enzymes were either lyophilized or directly (in liquid form)

stored at -80°C for further analytical studies.

Page 114: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

85

3.6. Characterization of Recombinant TnBglA and TnBglB

Characterization of recombinant TnBglA and TnBglB enzymes from T. naphthophila were

performed to determine molecular weight, optimal pH and temperature, thermal

inactivation, pH stability, substrate specificity, kinetics parameters, thermodynamics study

of substrate hydrolysis and enzymes inactivation, shelf life of enzymes, effect of chemical

inhibitors, metallic cation, glucose and xylose on the catalytic efficiency of cloned

enzymes.

3.6.1. Determination of Molecular weight

Purity and apparent molecular weight of recombinant TnBglA and TnBglB proteins were

analyzed by SDS-PAGE. Individually the purified TnBglA and TnBglB enzymes were

loaded on 12% SDS-gel along with control protein sample from both induced and

uninduced cells of E. coli BL21 CodonPlus harboring pET-21a(+) without insert for the

comparison of expressions and to determine the molecular weight of recombinant enzymes.

3.6.2. Optimal pH of TnBglA and TnBglB

To determine the optimal pH of purified recombinant TnBglA and TnBglB enzymes,

various buffers were used in the present study. For example, McIlvaine buffer (for pH 2.2-

8.0), sodium acetate buffer (for pH 3.7-5.6), Tris-Cl buffer (for pH 7.0-9.0), MES buffer

(for pH 5.6-6.6), Sorensen’s phosphate buffer (for pH 5.8-8.0), HEPES buffer (for pH 6.8–

8.2), MOP buffer (for pH 7.0-7.8), and CAPS buffer (for pH 9.0-11.0). To scrutinize the

optimum pH and buffer for TnBglA and TnBglB catalytic activity, the enzymes were

diluted suitably in buffers from pH 3.0-11.0, in increments of 0.5 unit of pH, under the

assay conditions as described earlier. The results were compiled as the percentage of

TnBglA and TnBglB activities.

3.6.3. Optimal Temperature of TnBglA and TnBglB

The optimal temperature of cloned TnBglA and TnBglB were measured over a broad

temperature ranged from 30-100°C, under standard assay conditions using already

investigated optimal pH buffer for both enzyme that was McIlvaine buffer having pH 7.0

Page 115: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

86

for TnBglA and pH 5.0 for TnBglB. Both enzymes were incubated for 10 minutes at

different temperatures (30-100°C), with the increments of 1 to 5 degree C followed by stop

the reactions with the addition of 3 mL 1 M Na2CO3. All results related to temperature

profile was expressed as percentage of TnBglA and TnBglB activities.

3.6.4. Optimal Incubation Time

Time course profile of recombinant enzymes (TnBglA and TnBglB) for substrate

hydrolysis were monitored to find the optimum incubation time for reaction under the

optimal assay conditions of TnBglA and TnBglB incubated at 95°C and 85°C using pH 7.0

and 5.0 buffer, respectively. Therefore, standard reaction mixtures of both enzymes were

incubated from 01 to 15 minutes subsequently computed the product released per minutes.

3.6.5. pH Stability

To evaluating the effect of H+ ions concentration on recombinant enzyme stability, pre-

incubated the purified enzymes (TnBglA and TnBglB) for 60 minutes in different buffers

ranging from pH 3.0-11.0 at 70°C, followed by diluted each sample by using McIlvaine

buffer (pH 7.0 for TnBglA and pH 5.0 for TnBglB) at 1:50 of dilution ratio and immediately

determined the residual activity of enzymes under optimal assay conditions for TnBglA

and TnBglB at 95°C (in pH 7.0 buffer) and 85°C (in pH 5.0 buffer), respectively. Results

were expressed as percentage of obtained residual activity, and enzyme activity defined as

100% which was without pre-incubation.

3.6.6. Thermal stability at different pH

Thermostability of recombinant purified TnBglA and TnBglB were investigated by

measuring the residual activity of pre-incubated aliquots at temperature ranging from 30-

100°C for different time intervals (up to 12 hours) in different buffer solutions ranging

from pH 4.0-7.5, in the absence of substrate. Aliquots were taken out followed by incubated

on ice for 30 minutes. Activity assay of TnBglA (95°C, pH 7.0) and TnBglB (85°C, pH

5.0) enzymes were performed under the standard assay conditions. Activity of TnBglA and

TnBglB without pre-incubation was considered as 100%.

Page 116: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

87

3.6.7. Effect of metallic ions and different inhibitors

To find out the inhibitory influence of various metallic cations on the activity of purified

TnBglA and TnBglB, enzymes were incubated in various metal cations (Mn2+, Ca2+, Zn2+,

Ni2+, Pb2+, Mg2+, Hg2+, Cu2+, Co2+, Cd2+ and Fe3+) at final concentration of 10 mM. The

effect of various chemical reagents and detergents (1% of β-mercaptoethanol, SDS, Triton-

X-100, Tween-80, and Urea) on the activity of recombinant enzymes were also

investigated. Chelating agent (10 mM EDTA) as well as different % (v/v) of short chain

alcohols (methanol, ethanol, n-butanol, and isopropanol) and organic solvents like ethyl

acetate, acetonitrile, acetone, dimethyl sulfoxide (DMSO) and Dimethylformamide (DMF)

were also used as chemical additives. The TnBglA and TnBglB residual activity were

measured using pNPG, under standard assay conditions. The activity of TnBglA and

TnBglB without any inhibitor, metallic ions and modifying agents was defined as 100%.

3.6.8. Effect of glucose and xylose

The effect of glucose and xylose on purified TnBglA and TnBglB enzyme activities were

investigated. The Ki value, representing the amount of glucose and xylose required to

inhibit activity of TnBglA and TnBglB up to 50% (Pei et al., 2012), was found by

incubating the purified recombinant enzyme in different concentrations of glucose and

xylose (20-1500 mM) and the residual activity of both purified enzymes were studied under

the standard assay conditions. Activity of enzymes without the addition of glucose and

xylose were considered as 100%.

3.6.9. Shelf life of recombinant TnBglA and TnBglB

The storage stability of recombinant TnBglA and TnBglB crude supernatant, crude after

heat treatment (for 1 h, at 70°C) and purified enzymes were monitored at room temperature

(30-35°C) and under refrigeration (4°C) conditions. Recombinant enzymes residual

activity were measured intermittently for a period of one year using standard assay

conditions as describe earlier.

Page 117: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

88

3.6.10. Substrate specificity

Substrate specificity of purified TnBglA and TnBglB were determined by using various

para- and ortho-nitrophenyl substrates such as pNP-β-D-galactopyranoside (pNPGal),

oNP-β-D-galactopyranoside (oNPGal), pNP-β-D-fucopyranoside (pNPF), oNP-β-D-

fucopyranoside (oNPF), pNP-β-D-xylopyranoside (pNPX), oNP-β-D-glucopyranoside

(oNPG), pNP-β-D-cellobioside (pNPC), pNP-α-D-glucopyranoside (pNPαG), pNP-α-L-

arabinofuranoside (pNPαA), pNP-β-D-mannoside (pNPM), pNP-β-D-glucuronide

(pNPGn) pNP-α-D-cellobioside (pNPαC) in the same way as with pNP-β-D-

glucopyranoside (pNPG, 4 mg mL-1) under the optimal assay conditions of TnBglA and

TnBglB.

When 1% cellobiose was used as substrate, the released amount of glucose was determined

using a glucose assay kit GAGO 20 (Sigma, Aldrich) according to the manufacturer’s

protocol. Glucose standard curve was prepared under the same assay conditions and was

used to find out the glucose concentration. Various polysaccharides substrates (4% CMC,

2% birchwood xylan, 2% beechwood xylan, 1% avicel, 1% sucrose and 0.5% laminarin)

were used in a 1 mL reaction mixture, under described assay conditions as in following

section.

3.6.11. Activity assay using polysaccharides

Various polysaccharides substrates (4% CMC, 2% birchwood xylan, 2% beechwood xylan,

avicel and laminarin) were used to scrutinize the catalytic activity, assays performed in seal

caped heat resistant tubes. Reaction mixture (1 mL) contained respective substrate (0.5

mL) in McIlvaine buffer (pH 7.0 and pH 5.0 accordingly) and diluted TnBglA (100 ng of

enzyme in 10 µL and 490 µL buffer, pH 7.0) or TnBglB (100 ng of enzyme in 10 µL and

490 µL buffer, pH 5.0). A single substrate assay with both enzymes were carried out in

triplicate along with a control reaction, followed by incubation at 95°C for TnBglA and at

85°C for TnBglB. After 10 minutes incubation, reaction was stopped using 3,5-

dinitrosalicylic acid (3 mL DNS) reagent, and mixture was incubated in a boiling water

bath for 10 minutes, measured absorbance against blank at 540 nm, and the concentration

Page 118: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

89

of liberated reducing sugars were quantified using DNS method described by Miller

(1959). The released reducing sugar was measured by using standard curve of glucose.

3.6.11.1. Standard Curve of Glucose and Xylose

To make the standard curve of glucose and xylose, stock solutions of glucose (50 mg mL-

1) and xylose (50 mg mL-1) were prepared in dH2O. Both glucose and xylose were diluted

serially ranging from 0.1-1.2 mg mL-1 in well dried label test tubes by using respective

stock solutions, and made final volume 1000 µL of the reaction. A control was also run

parallel by replacing glucose/xylose with dH2O, followed by incubation in boiling water

for 10 minutes, subsequently adding 3 mL of DNS in each serially diluted reaction tubes

and measured the absorbance at 540 nm using spectrophotometer. A standard curve of

glucose (Figure 3.8) and xylose were plotted by taking absorbance along Y-axis and

various concentration of glucose along X-axis (Figure 3.9).

Figure 3.8: Standard curve of Glucose.

y = 0.0073x + 0.0153R² = 0.9996

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

0 0.2 0.4 0.6 0.8 1 1.2 1.4

Ab

sorb

ance

(54

0 n

m)

Concentration mg mL-1

Standard Curve of Glucose

Page 119: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

90

Figure 3.9: Standard curve of Xylose.

3.6.11.2. Enzyme Unit

“One unit of recombinant enzyme activity is defined as the amount of enzyme that releases

1 µmol of reducing sugar per minute under the standard assay conditions.” Enzyme activity

in term of units was computed by applying the following formula:

3.6.11.3. Specific activity

Specific activity of TnBglA and TnBglB enzymes were described as enzyme units per mg

of the recombinant protein, and calculated by using the following formula:

3.6.12. Kinetic studies

To calculate Michaelis constant (Km), maximum velocity (Vmax) and enzyme turnover or

catalytic constants (kcat) Lineweaver-Burk plot was applied, various concentrations of

y = 0.0073x + 0.0153R² = 0.9996

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

0 0.2 0.4 0.6 0.8 1 1.2 1.4

Ab

sorb

an

ce (

54

0 n

m)

Concentration mg mL-1

Standard Curve of Xylose

Conc. from graph (mg mL-1) × Dil. factor × 1000

Incubation time (min) × MW of glucose/xylose Enzyme activity (U mL-1 min-1) =

U mL-1 min-1 of enzyme

mg of protein mL-1 Specific activity (U mg-1) =

Page 120: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

91

pNPG, pNPF, pNPGal, pNPX, pNPC and cellobiose substrates used with fixed amount of

recombinant enzymes. Rate of substrates hydrolysis for various concentration were

measured using McIlvaine buffer having enzyme optimal pH. Graphs were plotted by

taking 1/V along Y-axis and 1/[S] along X-axis, the values of Km and Vmax for all substrate

were calculated through Lineweaver-Burk plots and the value of kcat for each substrate was

computed by the following equation.

kcat = Vmax / [e]

Where, [e] is the concentration of enzyme.

3.6.13. Thermodynamics of substrate hydrolysis

Thermodynamics of pNPG hydrolysis were calculated as elucidated by Haq et al. (2012).

Activation energy (Ea) was calculated from the plot of ln Vmax against 1/T and

thermodynamic data (∆H*, ∆S* and ∆G*) were calculated by using the following

equations:

Ea (activation energy) = -slope × R

∆H* (enthalpy of activation) = Ea-RT

∆G* (free energy of activation) = -RT ln (kcath / kBT)

∆S* (entropy of inactivation) = (∆H* – ∆G*) / T

Where, ‘kB’ is the Boltzmann’s constant, kB is equal to the 1.3806488 × 10-23 J K-1, ‘T’ is

the absolute temperature in kelvin, ‘R’ is the gas constant which is equal to 8.3144621 J

K−1, and ‘h’ is the Planck’s constant which is equal to 6.62606957 x 10-34 J s or 11.04 x 10-

36 J. min.

3.6.14. Thermodynamics of TnBglA and TnBglB inactivation

Thermodynamic parameters for irreversible thermal denaturation of recombinant enzymes

were estimated by incubating purified enzyme at different temperatures for a specific time

period followed by determined the residual activity of enzymes. Thermal denaturation (Kd)

Page 121: A THESIS TITLED - prr.hec.gov.pk

Material & Methods

92

constant was calculated from a logarithmic plot of percent of remaining activity versus

time to determine the thermodynamic parameters for irreversible thermal denaturation of

recombinant TnBglA and TnBglB. EaD was calculated from plot of ln Kd against 1/T and

ΔH*D, ΔS*D and ΔG*D parameters were calculated using above mentioned equations after

replacing kcat and Ea with Kd and EaD, respectively as calculated by Haq et al. (2012).

Apparent half-lives (t1/2) of TnBglA and TnBglB were calculated using following equation.

t1/2 = In 2 / Kd

3.6.15. Statistical Analysis

All experimental reactions were conducted in CRD (completely randomized design) with

three replicates, data collection was statistically analyzed using student’s t-test analysis and

one-way ANOVA, with the significance results (p≤0.05) of ANOVA, Duncan’s multiple

range test was applied to analyze the comparison of means using SPSS software.

3.6.16. Docking Studies of Enzyme-Substrate

The minimized energy model structure of TnBglA and TnBglB were used for the docking

studies by PatchDock (Schneidman-Duhovny et al., 2005). Different substrates utilized in

the present study to test both enzymes activities, were also utilized for forecasting the

possible details of molecular enzyme substrate actions. The substrates docked with the both

β-glucosidases (TnBglA and TnBglB) included, cellobiose and p-nitrophenyl substrates.

All PatchDock results of docking were analyzed for evaluating the accuracy of docked

model and predicting the molecular details of both proteins in terms of non-covalent

interactions among enzyme (TnBglA and TnBglB) and substrates by using PDBsum

(Laskowski, 2009).

Page 122: A THESIS TITLED - prr.hec.gov.pk

CHAPTER-IV

RESULTS

Page 123: A THESIS TITLED - prr.hec.gov.pk

Results

93

4. RESULTS

The quest for thermotolerant and efficient cellulases led the way towards thermophiles.

The members of genus Thermotoga are believed to be an excellent source of highly active,

acidic-alkali resistant and highly heat-stable glycoside hydrolases (GHs) especially

cellulases. They are strictly anaerobes, rod-shaped, gram-negative bacteria which

optimally grow at 80-90°C. Therefore, in the present research work a marine

hyperthermophilic bacterium T. naphthophila RKU-10T (ATCC BAA-489/DSM

13996/JCM 10882) was selected for the isolation of novel cellulolytic genes because this

strain contains a large number of putative cellulolytic genes. Among these, two cellulolytic

genes were selected for molecular gene cloning and overexpression in mesophilic host E.

coli BL21 (DE3) followed by characterization and thermodynamic studies of both purified

enzymes were conducted.

All results related to the present study are discussed in the following section.

4.1. β-1,4-glucosidase genes from T. naphthophila

T. naphthophila RKU-10T complete genomic sequence was available on NCBI server

under GenBank accession number CP001839.1 and its genomic DNA has 1809823 base

pairs with several cellulolytic genes. Using the online NCBI database two putative

cellulolytic (β-1,4-glucosidase) genes fragments of 1,341 and 2,166 bp with locus tag

Tnap_0602 and Tnap_0656 were selected for the present research work and designated as

TnbglA and TnbglB. Gene bglA from T. naphthophila (TnbglA) encoding 446 amino acid

residues of a protein (TnBglA) and second gene bglB from T. naphthophila (TnbglB)

encoding 721 amino acid residues of a protein (TnBglB). Proteins TnBglA and TnBglB

have molecular weight (MW) of 51.50 kDa and 81.14 kDa, respectively.

All characteristics detail of both genes along with protein _ids from online NCBI database

software are presented in the following table 4.1.

Page 124: A THESIS TITLED - prr.hec.gov.pk

Results

94

Table 4.1: Characteristics details of TnbglA and TnbglB genes of T. naphthophila.

Property TnbglA TnbglB

Source Organism T. naphthophila RKU-10T T. naphthophila RKU-10T

Culture Collection ID DSM 13996, JCM 10882 DSM 13996, JCM 10882

Gene Size 1,341 bp 2,166 bp

Gene symbol TnbglA TnbglB

GenBank Accession no. CP001839.1 CP001839.1

Gene ID 281373136 281373190

Gene region 615833-617173 675125-677291

Locus tag Tnap_0602 Tnap_0656

Glycoside Hydrolase (GH)

family

GH family 1 (GH1) GH family 3 (GH3)

Protein_id ADA66698.1 ADA66752.1

4.2. Gene Sequence of TnbglA

The complete gene sequence of the putative cellulolytic gene belonged to GH family 1

was available on NCBI. Primers (forward and reverse) were designed using the obtained

gene sequences and the underline bold base pairs showed the NdeI restriction site

upstream (underlined) which was introduced to digest the gene with NdeI restriction

enzyme.

4.2.1. FASTA Sequence

“CATATGAACGTGAAAAAGTTCCCTGAAGGATTCCTCTGGGGTGTTGCAACAGCTTCCT

ACCAGATCGAGGGTTCTCCCCTCGCAGACGGAGCTGGTATGTCTATCTGGCACACCTTC

TCCCATACTCCTGGAAATGTAAAGAACGGTGACACGGGAGATGTGGCCTGCGACCACTA

CAACAGATGGAAAGAGGACATTGAAATCATAGAGAAACTCGGAGTAAAGGCTTACAGAT

TTTCAATCAGCTGGCCAAGAATACTTCCGGAAGGAACAGGAAGGGTGAATCAGAAAGGA

CTGGATTTTTACAACAGGATCATAGACACCCTGCTGGAAAAAGGTATCACACCCTTTGT

GACCATCTATCACTGGGATCTTCCCTTCGCTCTTCAGTTGAAAGGAGGATGGGCGAACA

GAGAAATAGCGGATTGGTTCGCAGAATACTCAAGGGTTCTCTTTGAAAATTTCGGCGAC

Page 125: A THESIS TITLED - prr.hec.gov.pk

Results

95

CGTGTGAAGAACTGGATCACCTTGAACGAACCGTGGGTTGTTGCCATAGTGGGGCATCT

GTACGGAGTCCACGCTCCTGGAATGAGAGATATTTACGTGGCTTTCCGAGCTGTTCACA

ATCTCTTGAGGGCACACGCCAAAGCGGTGAAAGTGTTCAGGGAAACTGTGAAAGATGGA

AAGATCGGAATAGTTTTCAACAATGGATATTTCGAACCTGCGAGTGAAAAAGAGGAGGA

CATCAGAGCGGCGAGATTCATGCATCAGTTCAACAACTATCCTCTCTTTCTCAATCCGA

TCTACAGAGGAGATTATCCGGAGCTCGTTCTGGAATTTGCCAGAGAGTATCTACCGGAG

AATTACAAAGATGACATGTCCGAGATACAGGAAAAGATCGACTTTGTTGGATTGAACTA

TTACTCCGGTCATTTGGTGAAGTTCGATCCAGATGCACCAGCTAAGGTCTCTTTCGTTG

AAAGGGATCTTCCAAAAACAGCCATGGGATGGGAGATCGTTCCAGAAGGAATCTACTGG

ATCCTGAAGAAGGTGAAAGAAGAATACAACCCACCAGAGGTTTACATCACAGAGAATGG

GGCTGCTTTTGACGACGTAGTTAGTGAAGATGGAAGAGTTCACGATCAAAACAGAATCG

ATTATTTGAAGGCCCACATTGGTCAGGCATGGAAGGCCATACAGGAGGGAGTGCCGCTT

AAAGGTTACTTCGTCTGGTCGCTCCTCGACAATTTCGAATGGGCAGAGGGATACTCTAA

GAGATTTGGTATTGTGTACGTGGACTACAGTACTCAAAAACGCATCATAAAAGACAGTG

GTTACTGGTACTCGAACGTGGTCAAAAGCAACAGTCTGGAAGATTGA”

4.2.2. Amino acid sequence of TnBglA

"MNVKKFPEGFLWGVATASYQIEGSPLADGAGMSIWHTFSHTPGNVKNGDTGDVACDHY

NRWKEDIEIIEKLGVKAYRFSISWPRILPEGTGRVNQKGLDFYNRIIDTLLEKGITPFV

TIYHWDLPFALQLKGGWANREIADWFAEYSRVLFENFGDRVKNWITLNEPWVVAIVGHL

YGVHAPGMRDIYVAFRAVHNLLRAHAKAVKVFRETVKDGKIGIVFNNGYFEPASEKEED

IRAARFMHQFNNYPLFLNPIYRGDYPELVLEFAREYLPENYKDDMSEIQEKIDFVGLNY

YSGHLVKFDPDAPAKVSFVERDLPKTAMGWEIVPEGIYWILKKVKEEYNPPEVYITENG

AAFDDVVSEDGRVHDQNRIDYLKAHIGQAWKAIQEGVPLKGYFVWSLLDNFEWAEGYSK

RFGIVYVDYSTQKRIIKDSGYWYSNVVKSNSLED”

4.3. Gene Sequence of TnbglB

The complete gene sequence of putative cellulolytic gene belonged to GH family 3 was

available on NCBI. Primers (forward and reverse) were designed using the obtained gene

sequences and the underline bold base pairs showed the Nde I restriction site upstream

(underlined) which was introduced to digest the gene with NdeI restriction enzyme.

Page 126: A THESIS TITLED - prr.hec.gov.pk

Results

96

4.3.1. FASTA Sequence

“CATATGGAAAGGATCGATGAAATCCTCTCTCAGTTAACTACAGAGGAAAAGGTGAAGC

TCGTTGTGGGGGTCGGTCTTCCAGGACTTTTTGGAAACCCACATTCCAGAGTGGCGGGT

GCGGCTGGAGAAACACATCCCATTCCAAGACTTGGAATTCCTGCGTTTGTCCTGGCAGA

TGGTCCCGCAGGACTCAGAATAAATCCCACAAGGGAAAACGATGAAAACACTTACTACA

CGACGGCATTTCCCGTTGAAATCATGCTTGCTTCTACCTGGAACAGAGACCTTCTGGAA

GAAGTGGGAAAAGCCATGGGAGAAGAAGTTAGGGAATACGGTGTCGATGTGCTTCTTGC

ACCTGCGATGAACATTCACAGAAACCCTCTTTGTGGAAGGAATTTCGAGTACTACTCAG

AAGATCCTGTCCTTTCCGGTGAAATGGCTTCAGCCTTTGTCAAGGGAGTTCAATCTCAA

GGGGTGGGAGCCTGCATAAAACACTTTGTCGCGAACAACCAGGAAACGAACAGGATGGT

AGTGGACACGATCGTGTCCGAGCGAGCCCTCAGAGAAATATATCTGAAAGGTTTTGAAA

TTGCCGTCAAGAAAGCAAGACCCTGGACCGTGATGAGCGCTTACAACAAACTGAATGGA

AAATACTGTTCACAGAACGAATGGCTTTTGAAGAAGGTTCTCAGGGAAGAATGGGGATT

TGACGGTTTCGTGATGAGCGACTGGTACGCGGGAGACAACCCTGTAGAACAGCTCAAGG

CCGGAAACGATATGATCATGCCTGGAAAAGCGTATCAGGTGAACACGGAAAGAAGAGAT

GAAATAGAAGAAATCATGGAGGCGTTGAAGGAGGGAAAGTTGAGTGAGGAGGTTCTCGA

TGAGTGTGTGAGAAACATTCTCAAAGTTCTTGTGAACGCGCCTTCCTTCAAAGGGTACA

GGTACTCAAACAAACCGGATCTCGAATCTCACGCGGAAGTCGCCTACAAAGCAGGTGCG

GAGGGTGTTGTCCTTCTTGAGAACAACGGTGTTCTTCCGTTCGATGAAAATACCCATGT

CGCCGTCTTTGGCACCGGTCAAATCGAAACAATAAAGGGAGGAACGGGAAGTGGAGACA

CCCATCCGAGATACACGATCTCTATCCTTGAAGGCATAAAAGAAAGAAACATGAAGTTC

GACGAAGAACTCGCTTCCACTTATGAGGAGTACATAAAAAAGATGAGAGAAACAGAGGA

ATATAAACCCAGAACCGACTCCTGGGGAACGGCCATAAAACCGAAACTTCCAGAGAACT

TCCTCCCAGAAAAAGAGATAAAGAAAGCTGCAAAGAAAAACGATGTTGCAGTTGTTGTG

ATCAGCAGGATCTCCGGTGAGGGATACGACAGAAAGCCGGTGAAAGGTGATTTCTACCT

CTCCGATGACGAGCTGGAACTCATAAAAACCGTCTCGAAAGAATTCCACGATCAGGGTA

AGAAAGTTGTGGTTCTTCTGAACATCGGAAGTCCCATCGAAGTCGCAAGCTGGAGAGAC

CTTGTGGATGGAATTCTTCTTGTCTGGCAGGCGGGACAGGAGATGGGAAGAATAGTGGC

CGATGTTCTTGTGGGAAAGATTAATCCCTCCGGAAAACTTCCAACGACCTTCCCGAAGG

ATTACTCGGACGTTCCATCCTGGACGTTCCCAGGAGAGCCAAAGGACAATCCGCAAAGA

GTGGTGTACGAGGAAGACATCTACGTGGGATACAGGTACTACGACACCTTTGGTGTGGA

Page 127: A THESIS TITLED - prr.hec.gov.pk

Results

97

ACCTGCCTACGAGTTCGGCTACGGCCTCTCTTACACAAAGTTTGAATACAAAGATTTAA

AGATCGCTATCGACGGAGATATACTCAGAGTGTCGTACACGATCACAAACACCGGGGAC

AGAGCTGGAAAGGAAGTCTCACAGGTTTATGTCAAAGCTCCAAAAGGGAAAATAGACAA

ACCCTTCCAGGAGCTGAAAGCGTTCCACAAAACAAAACTTTTGAACCCGGGTGAATCCG

AAAAGATCTTTCTGGAAATTCCTCTTAGAGATCTTGCGAGTTTCGATGGGAAAGAATGG

GTTGTCGAGTCAGGAGAATACGAGGTCAGGGTCGGTGCATCTTCGAGGGATATAAGGTT

GAAAGATGTTTTCTTGGTTGAAGAAGAAAAGAGATTCAAACCATGA”

4.3.2. Amino acid sequence of TnBglB

“MERIDEILSQLTTEEKVKLVVGVGLPGLFGNPHSRVAGAAGETHPIPRLGIPAFVLAD

GPAGLRINPTRENDENTYYTTAFPVEIMLASTWNRDLLEEVGKAMGEEVREYGVDVLLA

PAMNIHRNPLCGRNFEYYSEDPVLSGEMASAFVKGVQSQGVGACIKHFVANNQETNRMV

VDTIVSERALREIYLKGFEIAVKKARPWTVMSAYNKLNGKYCSQNEWLLKKVLREEWGF

DGFVMSDWYAGDNPVEQLKAGNDMIMPGKAYQVNTERRDEIEEIMEALKEGKLSEEVLD

ECVRNILKVLVNAPSFKGYRYSNKPDLESHAEVAYKAGAEGVVLLENNGVLPFDENTHV

AVFGTGQIETIKGGTGSGDTHPRYTISILEGIKERNMKFDEELASTYEEYIKKMRETEE

YKPRTDSWGTAIKPKLPENFLPEKEIKKAAKKNDVAVVVISRISGEGYDRKPVKGDFYL

SDDELELIKTVSKEFHDQGKKVVVLLNIGSPIEVASWRDLVDGILLVWQAGQEMGRIVA

DVLVGKINPSGKLPTTFPKDYSDVPSWTFPGEPKDNPQRVVYEEDIYVGYRYYDTFGVE

PAYEFGYGLSYTKFEYKDLKIAIDGDILRVSYTITNTGDRAGKEVSQVYVKAPKGKIDK

PFQELKAFHKTKLLNPGESEKIFLEIPLRDLASFDGKEWVVESGEYEVRVGASSRDIRL

KDVFLVEEEKRFKP”

4.4. Cloning and Sequencing of TnbglA and TnbglB Genes

Two cellulolytic (β-1,4-glucosidases) TnbglA and TnbglB genes fragment of 1,341 bp

(Tnap_0602) and 2,166 bp (Tnap_0656) from T. naphthophila RKU-10T, encoding 446

and 721 amino acid residues, respectively. Genomic DNA of T. naphthophila used as a

template for the amplification of β-1,4-glucosidase TnbglA and TnbglB genes, followed by

initially cloned in pTZ57R/T vector and after that sequencing were performed. All results

related to cloning in pTZ57R/T vector and sequencing analysis are described as below.

Page 128: A THESIS TITLED - prr.hec.gov.pk

Results

98

4.4.1. Determination of Non-cutter restriction enzymes

To identify all the non-cutter (zero-cutter) restriction enzymes for the TnbglA and TnbglB,

an online NEBcutter V2.0 software was used in order to introduce restriction site in the

forward primer. The software showed a linear sequence of target genes along with non-

cutter restriction enzymes.

4.4.2. Non-cutter enzymes for TnbglA

After uploading the FASTA nucleotide sequence, all restriction enzymes were displayed.

TnbglA contained 46% GC and 54% AT contents and encoding 446 amino acid residues.

Following figure 4.1a shows the single cutter of TnbglA gene and list of zero-cutters are

displays in the table 4.2a.

Figure 4.1 (a): Linear sequence of TnbglA gene with NEB single cutter endonucleases.

4.4.3. Non-cutter enzymes for TnbglB

TnbglB contained 47% GC and 53% AT contents and encoding 721 amino acid residues.

Following figure 4.1b shows the single cutter of TnbglB gene and list of zero-cutters are

displays in the table 4.2b.

Page 129: A THESIS TITLED - prr.hec.gov.pk

Results

99

Figure 4.1 (b): Linear sequence of TnbglB gene with NEB single cutter endonucleases.

Table 4.2 (a): List of Non-cutter enzymes for TnbglA.

AatII, Acc65I, AccI, AclI, AfeI, AflII, AgeI, AhdI, AleI, AlwNI, ApaI, ApaLI, AscI, AseI, AsiSI, AvaI, AvaII,

AvrII,

BaeI, BanI, BbsI, BbvCI, BceAI, BclI, BfaI, BglI, BglII, BlpI, BmgBI, BmtI, BpmI, BsaBI, BsaHI, BseYI,

BsgI, BsiWI, BsmBI, BsmFI, BsmI, BsoBI, BspCNI, BspHI, BsrDI, BsrFI, BsrGI, BssHII, BssSI, BstAPI,

BstEII, BstUI, BstZ17I, Bsu36I, BtgZI, BtsI,

CspCI,

DraI, DraIII, DrdI,

EagI, EciI, EcoO109I, EcoRI, EcoRV,

FauI, FseI, FspI,

HaeII, HgaI, HhaI, HinP1I, HincII, HindIII, HpaI,

KasI, KpnI,

MfeI, MluI, MslI,

NaeI, NarI, NciI, NdeI, NgoMIV, NheI, NmeAIII, NotI, NruI,

PacI, PaeR7I, PflFI, PluTI, PmeI, PmlI, PpuMI, PshAI, PsiI, PspOMI, PspXI, PstI, PvuI,

RsrII,

SacII, SalI, SbfI, SexAI, SfiI, SfoI, SgrAI, SmaI, SnaBI, SpeI, SphI, SrfI, SspI, StuI, SwaI

TspMI, Tth111I,

XbaI, XcmI, XhoI, XmaI,

ZraI

Page 130: A THESIS TITLED - prr.hec.gov.pk

Results

100

Table 4.2 (b): List of Non-cutter enzymes for TnbglB

AatII, Acc65I, AccI, AclI, AfeI, AflII, AgeI, AhdI, AleI, AlwNI, ApaI, ApaLI, AscI, AseI, AsiSI, AvaI, AvaII,

AvrII,

BaeI, BanI, BbsI, BbvCI, BceAI, BclI, BfaI, BglI, BglII, BlpI, BmgBI, BpmI, BsaBI, BsaHI, BseYI, BsgI,

BsiWI, BsmBI, BsmFI, BsmI, BsoBI, BspCNI, BspHI, BsrDI, BsrFI, BsrGI, BssHII, BssSI, BstAPI, BstEII,

BstUI, Bsu36I

CspCI,

DraI, DraIII, DrdI,

EagI, EciI, EcoO109I, EcoRI, EcoRV,

FauI, FseI, FspI,

HaeII, HgaI, HhaI, HinP1I, HincII, HindIII, HpaI,

KasI, KpnI,

MfeI, MluI, MslI,

NaeI, NarI, NciI, NdeI, NgoMIV, NheI, NmeAIII, NotI, NruI,

PacI, PaeR7I, PflFI, PluTI, PmeI, PmlI, PpuMI, PshAI, PsiI, PspOMI, PspXI, PstI, PvuI,

SacII, SalI, SbfI, SexAI, SfiI, SfoI, SgrAI, SmaI, SnaBI, SpeI, SphI, SrfI, SspI, StuI, SwaI,

TspMI, Tth111I,

XbaI, XcmI, XhoI, XmaI,

ZraI

4.4.4. Primers Designing

To amplify the coding regions of TnbglA (ADA66698.1) and TnbglB (ADA66752.1), two

sets of primers were designed using Vector NTI Advance TM 10.3 software (Invitrogen

Corporation, California) and OligoCalc software (Kibbe, 2007). NdeI restriction site

(CATATG) upstream (underlined) at 5' end of the both forward primers of TnbglA and

TnbglB genes were introduced. No restriction site was introduced in reverse primers

because in multiple cloning site of pTZ57R/T (cloning vector) has HindIII site (second

restriction endonuclease used for restriction digestion). The primers were used to amplify

Page 131: A THESIS TITLED - prr.hec.gov.pk

Results

101

TnbglA and TnbglB genes size of 1,341 and 2,166 bp, all thermodynamic properties and

sequences of designed oligomers (primers) are briefly described in the following Table 4.3.

Table 4.3: Oligonucleotides for PCR amplification of TnbglA and TnbglB genes.

Gene

Name Primer Sequence 5' 3'

R*

Site

Primer

length

(nt)

product

size

(bp)

MW Tm

TnbglA

F-CATATGAACGTGAAAAAGTTCCCTGAAGG

R-TCAATCTTCCAGACTGTTGCTTTTGACC

NdeI

-

29

28

1,341

8,959

8,481

61.2

61.0

TnbglB

F-CATATGGAAAGGATCGATGAAATCCTCTCTCA

R-TCATGGTTTGAATCTCTTTTCTTCTTCAACCAAG

NdeI

-

32

34

2,166

9,816

10,323

61.8

61.3

R* represented the restriction site, F (Forward), R (Reverse), MW (molecular weight), nt (nucleotides), bp

(base pairs), and Tm (melting temperature). TnbglA represented bglA gene and TnbglB represented bglB

gene from T. naphthophila.

Page 132: A THESIS TITLED - prr.hec.gov.pk

Results

102

4.4.5. Extraction of Genomic DNA

T. naphthophila was grown at 80°C for 166 hours in an anaerobic conditions followed by

the genomic DNA extracted from the bacterial culture using standard protocol. Total

extracted DNA was analyzed after agarose gel electrophoresis under UV light (Figure 4.2).

Concentration was determined by absorption measurement at λ260, DNA quantity was

estimated to be approximately 120-140 ng µL-1.

Figure 4.2: Agarose gel of isolated genomic DNA from Thermotoga naphthophila, 6 µg DNA was loaded

in Lanes 1-3 along with DNA Marker (M) #SM0333.

4.4.6. TnbglA and TnbglB Genes Amplification

Target β-1,4-glucosidases (TnbglA and TnbglB) genes fragments of 1.341 kb and 2.166 kb

from T. naphthophila RKU-10T were amplified using gene specific primers in a

thermocycler. PCR often generates point mutations, therefore all reactions were performed

at least in triplicate. After the completion of PCR, amplicons were analyzed using 0.8%

agarose electrophoresis, gene fragment of 1.341 kb and 2.166 kb were obtained (Figure

4.3a and 4.3b).

M 1 2 3

10

Genomic DNA

3

1

0.5

kb

Page 133: A THESIS TITLED - prr.hec.gov.pk

Results

103

Figure 4.3 (a): Analysis of TnbglA amplified product resolved on 0.8% agarose gel. Lane M, DNA ladder

# SM0333; Lane 1 and 2 showed PCR-amplified product of 1.341 kb.

Figure 4.3 (b): Analysis of TnbglB amplified product after agarose gel electrophoresis. Lane M, DNA

ladder # SM0333; Lane 1 and 2 showed PCR-amplified gene product of 2.166 kb length.

M 1 2

kb

10 8.0 6.0 5.0 4.03.0 2.52.0 1.5 1.2 1.00.90.8 0.7 0.6 0.5 0.4 0.30.20.1

1.341 kb

M 1 2 kb 10 8.0 6.0 5.0 4.03.0 2.52.0 1.5 1.2 1.00.90.8 0.7 0.6 0.5 0.4 0.3

2.166 kb

Page 134: A THESIS TITLED - prr.hec.gov.pk

Results

104

4.4.7. Purification of Amplicons

PCR amplicons of TnbglA and TnbglB yielded a single product of their required sizes,

excised the bands of both amplified DNA and subsequently extract from agarose gel using

QIA quick gel extraction kit. Gene clean products of PCR were electrophoresed and results

are shown in figure 4.4.

Figure 4.4: Analysis of TnbglA and TnbglB purified PCR product of 1.341 kb and 2.166 kb, respectively on

0.8% agarose gel after DNA gel extraction (gene clean) using Qiagen Quick gel extraction kit. Lane M, DNA

ladder # SM0333; Lane 1, displayed TnBglA purified product (1.341 kb); and Lane 2, showed purified PCR

TnbglB gene product (2.166 kb).

4.4.8. Cloning of genes in pTZ57R/T

Purified TnbglA (1.341 kb) and TnbglB (2.166 kb) genes product were cloned in linear

pTZ57R/T vector using TA cloning strategy as elucidated in materials and methods. After

the incubation of ligation reaction, recombinant pTZ57R/T–TnbglA and pTZ57R/T–

TnbglB thus obtained, which were used to transform competent cells of DH5α strain

followed by spread on individual LB-agar plates supplemented with X-gal, IPTG, and

ampicillin, kept all plates at 37°C for 16-18 hours. Since the cloning vector pTZ57R/T is

genetically marked with LacZ, bacterial transformants could be selected by blue/white

screening. High efficiency of transformation was observed because a fairly good number

of white (putative positive transformants) colonies appeared on both plates after overnight

incubation (Figure 4.5).

M 1 2

3.0

2.0 2.5

1.5

1.0

kb

1.341 kb

2.166 kb

Page 135: A THESIS TITLED - prr.hec.gov.pk

Results

105

Figure 4.5: After transformation of E. coli strain DH5α (competent cells) with ligation product of

pTZ57R/T–TnbglA and pTZ57R/T–TnbglB, several white and blue colonies were appeared on LB-agar

plates containging X-gal/IPTG/ampicilline. Putative recombinant (positive) well isolated white colonies were

selected by using blue-white sereening method.

4.4.9. Spotting of selected white colonies

Ten to sixteen well isolated putative recombinant white colonies harboring pTZ57R/T–

TnbglA and pTZ57R/T–TnbglB were selected (marked), duplically spotted or streak shortly

on freshly prepared LB-ampicillin agar plates and after that kept at 37°C incubator for

overnight, results were shown in the Figure 4.6.

Figure 4.6: Randamly selected well-isolated white colonies from master plates (TnbglA and TnbglB) and

made short streak or duplically spotted on freshly prepared LB-ampicillin agar plates.

TnbglA TnbglB

TnbglA TnbglB

Page 136: A THESIS TITLED - prr.hec.gov.pk

Results

106

4.4.10. Colony PCR

By using gene-specific primers, the presence of insert genes (TnbglA and TnbglB) were

confirmed by colony PCR. Ten putative white colonies were screened for TnbglA and

thirteen colonies for TnbglB from spotted plates, and performed colony PCR followed by

gel electrophoresis to visualized the result as the described method (Figure 4.7a and 4.7b).

Figure 4.7 (a): Screening of positive transformants of E. coli DH5α harboring pTZ57R/T–TnbglA plasmids

by colony-PCR. Lane M, DNA ladder; Lane 1-10, amplified products of randomly selected white colonies;

Lane C, control reaction product. All selected white colonies were positive for TnbglA gene, arrow indicated

the position of 1.341 kb amplicon.

Figure 4.7 (b): Screening of positive transformants of E. coli DH5α harboring pTZ57R/T–TnbglB by colony-

PCR. Lane M, DNA size marker; Lane 1-13, amplified products of different selected colonies (PCR

reactions); Lane C, control reaction. All selected colonies were positive except colony no. 11 which was

negative for TnbglB insert. The arrow specified the position of 2.166 kb amplicon.

1.5

2.0

0.5

1.0

1.2

3.0

4.0

5.0

M 1 2 3 4 5 6 7 8 9 10 C

1.341 kb

kb

2.5 2.0

M 1 2 3 4 5 6 7 8 9 10 11 12 13 C

kb

8.0 3.0

2.166 kb

1.0

0.2 0.5

Page 137: A THESIS TITLED - prr.hec.gov.pk

Results

107

4.4.11. Extraction of Recombinant Plasmids

After analyzing the results of colony PCR of both genes, recombinant plasmids

(pTZ57R/T–TnbglA and pTZ57R/T–TnbglB) were extracted from four positive bacterial

transformants (encircled) by alkaline lysis method and treated with RNase (Figure 4.8a and

4.8b) for further restriction analysis and sequencing.

Figure 4.8: Isolated recombinant plasmids pTZ57R/T–TnbglA and pTZ57R/T–TnbglB from four to five

selected positive transformants of E. coli DH5α after the screening by colony-PCR of both genes. (a) Lane

M, DNA ladder; Lane 1-5, recombinant uncut pTZ57R/T–TnbglA plasmids (circular) from five different

colonies after RNase treatment. (b) Lane M, DNA ladder; Lane 1-4, recombinant pTZ57R/T–TnbglB

plasmids (circular/uncut) from four different colonies after RNase treatment.

4.4.12. Single Restriction Analysis of Recombinant Plasmids

Plasmids (pTZ57R/T–TnbglA and pTZ57R/T–TnbglB) were used to confirm the presence

of insert genes by single restriction analysis with specific restriction enzymes, NdeI or

HindIII. Restriction analysis of both inserts were performed individually. DNA restriction

pattern of both recombinant plasmids were analyzed under UV-light after electrophoresed

on 0.8% agarose gel along with DNA size marker (Figure 4.9a and 4.9b).

kb

M 1 2 3 4 5 M M 1 2 3 4

(a) (b)

6.0 3.0 1.0

Uncut

Plasmids

Page 138: A THESIS TITLED - prr.hec.gov.pk

Results

108

Figure 4.9 (a): Restriction analysis of pTZ57R/T–TnbglA plasmid using single restriction enzyme. Lane M,

DNA ladder; Lane 1-4, restricted plasmids with HindIII from four different selected colonies and arrow

specified resultant bands of ~4.227 kb (2.886 kb pTZ+1.341 kb insert) for linearized pTZ57R/T–TnbglA.

Figure 4.9 (b): Restriction analysis of pTZ57R/T–TnbglB plasmid using single restriction enzyme. Lane M,

DNA ladder; Lane 1-4, restricted plasmids with HindIII from four selected colonies and resultant bands of

~5.052 kb (2.886 kb pTZ+2.166 kb insert) showed linear pTZ57R/T–TnbglB plasmid.

4.4.13. Sequence Analysis of TnbglA and TnbglB

Isolated recombinant plasmids (pTZ57R/T–TnbglA and pTZ57R/T–TnbglB) from four

selected positive transformants (encircled) were subjected to sequence analysis, both in

forward and reverse direction. The complete TnbglA and TnbglB sequences were identical

for all cloned. Nucleotide and amino acid sequences were analyzed (aligned) to the

kb

~ 4.227kb 4.0

8.0

3.0

1.5

1.0

0.5

M 1 2 3 4

M 1 2 3 4

kb

~ 5.052 kb

6.0

3.0

5.0

4.0

Page 139: A THESIS TITLED - prr.hec.gov.pk

Results

109

sequences available in GenBank database by utilizing Blast server at NCBI and Clustal

Omega.

4.4.14. Nucleotide Sequences Comparison of TnbglA gene

The nucleotide query sequence of TnbglA gene was blasted to identify and compare the

gene sequence with highly similar sequences in the NCBI database. Query produced

significant alignments after run and majority aligned sequences were β-glucosidases of

hyperthermophilic bacteria especially belonging to the genus Thermotoga and other

thermophiles. The query sequence of TnbglA showed 100% identity to the sequence of

Thermotoga naphthophila RKU-10T (CP001839.1) with 100% query coverage (Figure

4.10a). Further, the query sequence had high similarity of 99%, 99%, 99%, 99%, 98%,

98%, 82% and 82% to β-glucosidases from Thermotoga sp. RQ2, Thermotoga sp. Cell2,

Thermotoga sp. 2812B, T. petrophila RKU-1, T. maritima strain Tma100, T. maritima

MSB8, T. neapolitana DSM 4359 and Thermotoga sp. RQ7, respectively with 100% query

coverage. However, TnbglA nucleotide sequence showed 68%, 68%, 68%, 68% and 67%

similarity to β-glucosidases from Petrotoga mobilis SJ95, Clostridium thermocellum,

Ruminiclostridium thermocellum DSM 2360, Fervidobacterium pennivorans strain DYC

and Defluviitoga tunisiensis, respectively. Genomic sequence analysis revealed that β-1,4-

glucosidase TnbglA belongs to GH family 1. The eight significant aligned sequences are

presented in table 4.4a.

BLAST Result

Score Expect Identities Gaps Strand

2477 bits(1341) 0.0 1341/1341(100%) 0/1341(0%) Plus/Minus

Page 140: A THESIS TITLED - prr.hec.gov.pk

Results

110

Page 141: A THESIS TITLED - prr.hec.gov.pk

Results

111

Figure 4.10 (a): Sequence alignment between cloned TnbglA and NCBI retrieved sequence. Query = Cloned

TnbglA sequence, Subject = retrieved sequence from NCBI.

Table 4.4 (a): Nucleotide sequence BLAST result of TnbglA.

Description Accession

No.

Genomic

length

Query

Cover Score

E

value Gaps

Max.

Identity

Thermotoga sp. RQ2 CP000969.1 1877693 100% 2444 bits

(1323) 0.0

0/1341

(0%)

1335/134

(99%)

T. petrophila RKU-1 CP000702.1 1823511 100% 2466 bits

(1335) 0.0

0/1341

(0%)

1339/134

(99%)

Thermotoga sp. Cell2 CP003409.1 1749904 100% 2438 bits

(1320) 0.0

0/1341

(0%)

1334/134

(99%)

Thermotoga sp. 2812B CP003408.1 1843731 100% 2372 bits

(1284) 0.0

0/1341

(0%)

1322/134

(99%)

T. maritima MSB8 CP011107.1 1869610 100% 2361 bits

(1278) 0.0

0/1341

(0%)

1320/134

(98%)

T. neapolitana DSM 4359 CP000916.1 1884562 100% 1344 bits

(1490) 0.0

4/1343

(0%)

1105/134

3 (82%)

Defluviitoga tunisiensis LN824141.1 2053097 96% 356 bits

(394) 8e-94

34/131

5 (2%)

877/1315

(67%)

Fervidobacterium

pennivorans CP003260.1 2166381 53%

109 bits

(120) 2e-19

5/368

(1%)

248/368

(67%)

Page 142: A THESIS TITLED - prr.hec.gov.pk

Results

112

4.4.15. Nucleotide Sequences Comparison of TnbglB gene

The recombinant pTZR/T57–TnbglB gene was sequenced and FASTA nucleotide query

sequence of TnbglB gene was blasted to identify and compare the gene sequence with

highly similar sequences in the NCBI database. The query sequence of TnbglB showed

100% identity to the sequence of Thermotoga naphthophila RKU-10 (CP001839.1) with

100% query coverage (Figure 4.10b).Query produced significant alignments after run and

majority aligned sequences were beta-glucosidases of hyperthermophilic bacteria

especially belonged to the genus Thermotoga and other thermophiles. The query sequence

of TnbglB had high similarity of 99%, 99%, 98%, 98%, 95%, 80%, 79%, 73% and 67% to

β-glucosidases of GH family 3 from Thermotoga sp. RQ2, Thermotoga sp. Cell2,

Thermotoga sp. 2812B, T. maritima MSB8, T. petrophila RKU-1, Thermotoga sp. RQ7, T.

neapolitana DSM 4359, T. hypogea DSM 11164 and Fervidobacterium islandicum,

respectively. Genomic sequence analysis revealed that β-1,4-glucosidase TnbglB belongs

to GH family 3. The nine significant aligned sequences are presented in table 4.4b.

BLAST Result

Score Expect Identities Gaps Strand

4000 bits(2166) 0.0 2166/2166 (100%) 0/2166 (0%) Plus/Minus

Page 143: A THESIS TITLED - prr.hec.gov.pk

Results

113

Page 144: A THESIS TITLED - prr.hec.gov.pk

Results

114

Figure 4.10 (b): Sequence alignment between cloned TnbglB and NCBI retrieved sequence. Query =

Cloned TnbglA sequence, Subject = retrieved sequence from NCBI.

Page 145: A THESIS TITLED - prr.hec.gov.pk

Results

115

Table 4.4 (b): Nucleotide sequence BLAST result of TnbglB.

Description Accession

No.

Genomic

length

Query

Cover Score

E

value Gaps

Max.

Identity

Thermotoga sp. RQ2 CP000969.1 1877693 100%

3956 bits

(2142) 0.0

0/2166

(0%)

2158/2166

(99%)

Thermotoga sp. Cell2 CP003409.1 1749904 100%

3921 bits

(2123) 0.0

1/2166

(0%)

2152/2166

(99%)

Thermotoga sp. 2812B CP003408.1 1843731 100%

3757 bits

(2034) 0.0

0/2166

(0%)

2122/2166

(98%)

T. maritima MSB8 CP004077.1 1869612 100%

3746 bits

(2028) 0.0

0/2166

(0%)

2120/2166

(98%)

T. petrophila RKU-1 CP000702.1 1823511 99%

3374 bits

(1827) 0.0

1/2164

(0%)

2052/2164

(95%)

T. neapolitana DSM

4359 CP000916.1 1884562 97%

1576 bits

(853) 0.0

22/2127

(1%)

1706/212

(79%)

T. hypogea DSM

11164 CP007141.1 2165416 97%

1196 bits

(1326) 0.0

22/2122

(1%)

1548/2122

(73%)

T. caldifontis AZM44 AP014509.1 2014912 96%

1135 bits

(1258) 0.0

28/2115

(1%)

1529/2115

(72%)

Fervidobacterium

islandicum CP014334.1 2237377 94%

627 bits

(694) 5e-175

43/2056

(2%)

1386/2056

(67%)

4.4.16. Multiple sequence alignment of TnBglA and TnBglB

4.4.16.1. Alignment of TnBglA

Clustal omega program is used for the multiple sequence alignment of TnBglA to the others

members of GH1 from Genbank database, which exhibited high amino acid sequence

homology with β-glucosidases of genus Thermotoga especially with T. petrophila RKU-1

and T. maritima with 99% and 98% similarity, respectively. According to the homology

and structure modeling study, TnBglA has conserved structural folds and catalytic cleft for

substrate, with two or at the highest three conserved acidic amino acids residues (Glu166,

Glu351 and Glu405) present in the catalytic cleft or active site, one Glu residue in N-terminal

Page 146: A THESIS TITLED - prr.hec.gov.pk

Results

116

and other in middle or C-terminal position, play role in hydrolysis (Figure 4.11a), which

presented the low evolutionary pressure on these catalytic regions.

Alignment of β-glucosidase A from T. naphthophila (TnBglA)

T. naphthophila -MNVKKFPEGFLWGVATASYQIEGSPLADGAGMSIWHTFSHTPGNVKNGDTGDVACDHY 58 T. maritima ---MKKFPEGFLWGVATASYQIEGSPLADGAGMSIWHTFSHTPGNVKNGDTGDVACDHY 56 T. pseudethanolicus ----MIKFPKDFLWGTATSSYQIEGAVNEDGRTPSIWDTFSKTEGKTYNGHTGDVACDHY 56 T. ethanolicus ---MEVKFPKDFLWGTATSSYQIEGAVNEDGRTPSIWDTFSKTEGKTYNGHTGDVACDHY 57 C. staminisolvens --MSKINFPKDFIWGSATAAYQIEGAYNEDGKGESIWDRFSHTPGNIENGHNGDIACDHY 58 S. griseoaurantiacus MTIDLGAFPRDFLWGTATAAYQIEGAAAEDGRSPSIWDTYSHTPGKVAGGDHGDVACDHY 60 N. alba -MSKPDDFPEDFLWGAATASFQIEGATTADGRGRSIWDTFAETPGKVLNGDTGDPADDHY 59 T. naphthophila NRWKEDIEIIEKLGVKAYRFSISWPRILPEGTGRVNQKGLDFYNRIIDTLLEKGITPFVT 118 T. maritima NRWKEDIEIIEKLGVKAYRFSISWPRILPEGTGRVNQKGLDFYNRIIDTLLEKGITPFVT 116 T. pseudethanolicus HRYKEDVEILKEIGVKAYRFSIAWPRIFPE-EGKYNPKGMDFYKRLIDELLKKDIMPTAT 115 T. ethanolicus HRYKEDVEILKEIGVKAYRFSIAWPRIFPE-EGKYNSKGMDFYKRLVDELLKKDIMPTAT 116 C. staminisolvens HRYEEDIKIMKEIGLKSYRFSISWPRIFPEGTGQLNQKGLDFYKRLTNMLLENGIMPAIT 118 S. griseoaurantiacus HRWEEDIELMRRLGTNAYRLSVAWPRVVPGGTGEVNAKGLDFYDRLIDALLAAGITPSVT 120 N. alba HRYAEDIGLMRALNLGAYRFSIAWPRILPEGEGKVNQAGLDFYDRLVDALLEAGIRPWAT 119 * T. naphthophila IYHWDLPFALQLKGG-WANREIADWFAEYSRVLFENFGDRVKNWITLNEPWVVAIVGHLY 177 T. maritima IYHWDLPFALQLKGG-WANREIADWFAEYSRVLFENFGDRVKNWITLNEPWVVAIVGHLY 175 T. pseudethanolicus IYHWDLPQWAYDKGGGWLNRDSVKWYVEYATKLFEELGDVIPLWITHNEPWCSSILSYGI 175 T. ethanolicus IYHWDLPQWAYDKGGGWLNRDSVKWYVEYATKLFEELGDVIPLWITHNEPWCASILSYGI 176 C. staminisolvens LYHWDLPQKLQDKGG-WKNRDTTDYFTEYCEAIFKSLGDIVPMWITHNEPRVVALLGHFL 177 S. griseoaurantiacus LYHWDLPQVLQDRGG-WPERDTALAFASYAGVVAERLGDRVKMWTTLNEPLCSAWIGHLE 179 N. alba LYHWDLPQPLEDRGG-WPERDTALRFADYATVVAEALGDRVGDWMTINEPWCSAFLGYDN 178 T. naphthophila GVHAPGMRDIYVAFRAVHNLLRAHAKAVKVFRETVKD-GKIGIVFNNGYFEPASEKEEDI 236 T. maritima GVHAPGMRDIYVAFRAVHNLLRAHARAVKVFRETVKD-GKIGIVFNNGYFEPASEKEEDI 234 T. pseudethanolicus GEHAPGHKNYREALIAAHHILLSHGEAVKAFREMNIKGSKIGITLNLTPAYPASEKEEDK 235 T. ethanolicus GEHAPGHKNYREALIAAHHILLSHGEAVKAFREMNIKGSKIGITLNLTPAYPASEKEEDK 236 C. staminisolvens GIHAPGIKDLRTSLEVSHNILLSHGKTVKLFREMNID-AQIGIALNLSHHYPVSEKPEDI 236 S. griseoaurantiacus GLMAPGLTDLTAAVRASYHLLLGHGLAVRAVRAAVPD-AKVGIVNNLAWVEPATDRPEDV 238 N. alba GHHAPGRRDTAAALAATHHLLLGHGLAVEAIRSTGHP-ARVGLAHNQAVIRANGAHAADV 237 T. naphthophila RAARFMHQFNNYPLFLNPIYRGDYPELVLEFAREYLPEN---YKDDMSEIQEKIDFVGLN 293 T. maritima RAVRFMHQFNNYPLFLNPIYRGDYPELVLEFAREYLPEN---YKDDMSEIQEKIDFVGLN 291 T. pseudethanolicus LAAQYADGFANR-WFLDPIFKGNYPEDMMELYSKIIGEFDFIKEGDLETISVPIDFLGVN 294 T. ethanolicus LAAQYADGFANR-WFLDPIFKGNYPEDMMELYSKIIGEFDFIKEGDLKTISVPIDFLGVN 295 C. staminisolvens AAAELSFSIAGR-WYLDPLFKGCYPEDALEYYKKKGIELSFPQD-DLKLISQPIDFLAFN 294 S. griseoaurantiacus AAARRMDGHSNR-WWLDPVHGRGFPED-MREVYG-VELP--ERAGDLAAMAEPLDWLGLN 293 N. alba RAARRADGVRNR-IFTDPLFKGSYPADVLKDIAH-ISDFSFVREGDLATISAPIDFLGVN 295 T. naphthophila YYSGHLVKFD----------------PDAPAKVSFVERDLPKTAMGWEIVPEGIYWILKK 337 T. maritima YYSGHLVKFD----------------PDAPAKVSFVERDLPKTAMGWEIVPEGIYWILKK 335 T. pseudethanolicus YYTRSIVKYN----------------EDSMLKAENVPGPGKRTEMGWEISPESLYDLLKR 338 T. ethanolicus YYTRSIVKYN----------------EDSMLKAENVPGPGKRTEMGWEISPESLYDLLKR 339 C. staminisolvens NYSSDIIQYDPSD-------------ESGFSFAESILDKFEKTDMGWIIYPEGLYDLLML 341 S. griseoaurantiacus YYFPSVVED-----DPAG--------PAPRARAVRR-PGLPRTGMDWEVDAHGIERLLLR 339 N. alba YYTPEFVAGSDRGLDPAGVDGGGGAWPGAEPEEVHVSQGLPVTQMGWEIDPRGLYDVLQR 355 * T. naphthophila VKEEYNPPEVYITENGAAFDDVVSEDGRVHDQNRIDYLKAHIGQAWKAIQEGVPLKGYFV 397 T. maritima VKEEYNPPEVYITENGAAFDDVVSEDGRVHDQNRIDYLKAHIGQAWKAIQEGVPLKGYFV 395 T. pseudethanolicus LDREYTKLPMYITENGAAFKDEVTEDGRVHDDERIEYIKEHLKAAAKFIGEGGNLKGYFV 398 T. ethanolicus LDREYTKLPMYITENGAAFKDEVTEDGRVHDDERIEYIKEHLKAAAKFIGERGNLKGYFV 399 C. staminisolvens LDRDYGKPNLIISENGAAFKDEISSSGRIEDTKRIQYLKSYLTQAHRAIQDGVNLKGYYL 401 S. griseoaurantiacus LTEDYGARRLYITENGSAYPDVVGPDGTVDDPERTAYLEEHLAACASAVRKGVPLAGYFA 399 N. alba LAGESGGIDLYVTENGCAFEDRVSEDGAVHDPERTDYYEAHLRAAREAVHAGIPLRGYFA 415

Page 147: A THESIS TITLED - prr.hec.gov.pk

Results

117

* T. naphthophila WSLLDNFEWAEGYSKRFGIVYVDYSTQKRIIKDSGYWYSNVVKSNSLED- 446 T. maritima WSLLDNFEWAEGYSKRFGIVYVDYSTQKRIVKDSGYWYSNVVKNNGLED- 444 T. pseudethanolicus WSLMDNFEWAHGYSKRFGIVYVDYETQKRILKDSALWYKEVIQKNSIE— 446 T. ethanolicus WSLMDNFEWAHGYSKRFGIVYVDYETQKRILKDSALWYKEVIQKNSMS— 447 C. staminisolvens WSLMDNFEWSFGYNKRFGIVHVNFETQERKIKDSGYWYKEVIKNNGF--- 448 S. griseoaurantiacus WSLLDNFEWAYGYEKRFGLVHVDYATQRRTIKGSGHRYAELIRAHREPKA 449 N. alba WSLLDNFEWAWGYSRRFGIVYVDYETQERVVKDTGHWYAELAGTGRFPER 465

Figure 4.11 (a): Multi-alignment of β-glucosidase from T. naphthophila (TnBglA) with some other GH1

family β-glucosidases. Highly conserved amino acid residues are indicated by blocks. The catalytic sites are

represented by asterisk (*) on the top of alignment, the conserved catalytic Glu residues which act as proton

donor (E166) and nucleophile active site (E351 and E405). Thermotoga naphthophila RKU-10 (ADA66698.1),

Thermotoga maritima (WP_004082398.1), Clostridium straminisolvens (WP_038286797.1),

Thermoanaerobacter pseudethanolicus ATCC 33223 (ABY95558.1), Thermoanaerobacter ethanolicus

(WP_003869870.1), Streptomyces griseoaurantiacus (WP_006139294.1), Nocardiopsis alba

(WP_014910366.1).

4.4.16.2. Alignment of TnBglB

Clustal omega (ClustalW2) program is used for the multiple sequence alignment of TnBglB

amino acid sequence to the others members of thermostable GH3 from Genbank database,

which exhibited high amino acid sequence homology with β-glucosidases of genus

Thermotoga especially with T. petrophila RKU-1 and T. maritima with 99% and 98%

similarity, respectively. According to the homology and structure modeling study, TnBglB

has conserved structural folds and catalytic cleft for substrate, with two conserved acidic

amino acids residues (Asp242 and Glu458) present in conserved regions of amino acid

residues and formed the catalytic cleft or active site, which played hydrolytic role (Figure

4.11b). The conserved regions represented the low evolutionary pressure on these regions.

Alignment of β-glucosidase B from T. naphthophila (TnBglB)

Thermotoga naphthophila -MERIDEILSQLTTEEKVKLVVGVGLPGLFGNPHSRVAGAAGETHPIPRL 49 Thermotoga petrophila MMGKIDEILSQLTIEEKVKLVVGVGLPGLFGNPHSRVAGAAGETHPVPRL 50 Thermosipho africanus ----IEKIISQMTVEEKLKLLVGVGLPGMFGNKSSRVPGAAGETHQIERL 46 Fervidobacterium nodosum -MLDIEKVISQMTLEEKIHFVVGVGVLGIEDNPKARVSGAAGETFEIPRL 49 Thermotoga maritima -MERIDEILSQLTTEEKVKLVVGVGLPGLFGNPHSRVAGAAGETHPVPRL 49 Thermotoga neapolitana -MEKVNEILSQLTLEEKVKLVVGVGLPGLFGNPHSRVAGAAGETHPVPRV 49 Thermofilum sp. ----IEELLSKLSIEEKVKILVGIG--DTTSSQLARVPGAAGQTHPIERL 44 Thermofilum pendens -----------LSLEEKASLVVGWG------S-SRRLPGAAGETRPVR-- 30 Caldivirga maquilingensis --------VEDLSLEERVQLLVGAS------WRLRRIHGTAGETRPVRG- 35

Page 148: A THESIS TITLED - prr.hec.gov.pk

Results

118

Thermotoga naphthophila GIPAFVLADGPAGLRINPTRENDENTYYTTAFPVEIMLASTWNRDLLEEV 99 Thermotoga petrophila GIPSFVLADGPAGLRINPTRENDENTYYTTAFPVEIMLASTWNKDLLEEV 100 Thermosipho africanus KIPSTVHADGPAGLRIDPERENDSNKYHATAFPIASMLASTWNKEILFEV 96 Fervidobacterium nodosum GIPRTVYADGPAGLRIDPERESDERKYHATAFPVETMLASTWNKNILKKV 99 Thermotoga maritima GIPAFVLADGPAGLRINPTRENDENTYYTTAFPVEIMLASTWNRDLLEEV 99 Thermotoga neapolitana GLPAFVLADGPAGLRINPTRENDENTYYTTAFPVEIMLASTWNRELLEEV 99 Thermofilum sp. NIPGFVLADGPAGVRIEN------PACRATAFPVEVMLASTWNPELVEQV 88 Thermofilum pendens -VPSIVTADGPSGLRVEP---SGGRRWFATAFPVPTMLAATWNPEVVERV 76 Caldivirga maquilingensis -LPMVAMADGPSGIRIEP---NPIRRWPATAFPVPTMLASTWNPELVEAV 81 Thermotoga naphthophila GKAMGEEVREYGVDVLLAPAMNIHRNPLCGRNFEYYSEDPVLSGEMASAF 149 Thermotoga petrophila GKAMGEEVREYGVDVLLAPAMNIHRNPLCGRNFEYYSEDPVLSGEMASAF 150 Thermosipho africanus GKAMGNEAKEYGVDFLLAPAINIHRNPLCGRNFEYYSEDPILTGELASAF 146 Fervidobacterium nodosum GEAMGEEVREYGVDVLLAPAINIHRNPLCGRNFEYYSEDPLLTGELAASF 149 Thermotoga maritima GKAMGEEVREYGVDVLLAPAMNIHRNPLCGRNFEYYSEDPVLSGEMASAF 149 Thermotoga neapolitana GKAMGEEVREYGVDVLLAPAMNIHRNPLCGRNFEYYSEDPVLSGEMASSF 149 Thermofilum sp. GKAMGEEARSCGVDVLLAPALNIHRHPLGGRNFEYFSEDPLLSGKIASAY 138 Thermofilum pendens GRAIGEECRAYGVDVLLAPGVNMHRHPLCGRNFEYFSEDPLLSGEMAAAY 126 Caldivirga maquilingensis GRAMGEEARDYGIGVFLAPGINIHRHPLCGRNFEYFSEDPLLAGKIASAY 131 Thermotoga naphthophila VKGVQSQGVGACIKHFVANNQETNRMVVDTIVSERALREIYLKGFEIAVK 199 Thermotoga petrophila VKGVQSQGVGACIKHFVANNQETNRMVVDTIVSERALREIYLKGFEIAVK 200 Thermosipho africanus VEGVQSEGIGTSLKHFAANNQETNRMKIDTIVSERALREIYLKGFEIVVK 196 Fervidobacterium nodosum VEGVQSQGVGACLKHFVVNEQETNRMTVDTIVSERALREIYLKPFEIAIK 199 Thermotoga maritima VKGVQSQGVGACIKHFVANNQETNRMVVDTIVSERALREIYLKGFEIAVK 199 Thermotoga neapolitana VKGVQSQGVGACIKHFVANNQETNRMVVDTIVSERALREIYLRGFEIAVK 199 Thermofilum sp. VRGVQSAGIGACIKHFVGNEQETGRWGLDTFVSERALREIYLKPFEIAVK 188 Thermofilum pendens VRGVQSVGVGATLKHFAANDQETNRTVIDTVVSERALREIYLKPFEIAVK 176 Caldivirga maquilingensis VKGVQSVGVAATPKHFAANEQETNRTTVDTIVDERTLREIYLKPFEIVVK 181

* Thermotoga naphthophila KARPWTVMSAYNKLNGKYCSQNEWLLKKVLREEWGFDGFVMSDWYAGDNP 249 Thermotoga petrophila KARPWTVMSAYNKLNGKYCSQNEWLLKKVLREEWGFDGFVMSDWYAGDNP 250 Thermosipho africanus KAKPWTVMSAYNKLNGKYCSQNKWLLTKVLREEWGFEGFVMSDWFAGDNP 246 Fervidobacterium nodosum KSKPWTVMSSYNKLNGYYTSQNSWLLTKVLRYEWQFDGFVMTDWFAGDDG 249 Thermotoga maritima KARPWTVMSAYNKLNGKYCSQNEWLLKKVLREEWGFDGFVMSDWYAGDNP 249 Thermotoga neapolitana KSKPWSVMSAYNKLNGKYCSQNEWLLKKVLREEWGFEGFVMSDWYAGDNP 249 Thermofilum sp. EAKPWSVMSAYNKLNGVHCSENEWLLTTVLREEWGFDGFVMTDWGAGEDV 238 Thermofilum pendens KAKPWCVMSSYNKLNGKYSSQNEWLLTRVLREEWGFDGVVMTDWGAGDDS 226 Caldivirga maquilingensis EAKPWAIMSSYNKLNGKYASQNEWLLTKVLREEWGFDGIVMSDWGAGDNP 231 Thermotoga naphthophila VEQLKAGNDMIMPGKAYQVNTERRDEIEEIMEALKEGKLSEEVLDECVRN 299 Thermotoga petrophila VEQLKAGNDMIMPGKAYQVNTERRDEIEEIMEALKEGRLSEEVLNECVRN 300 Thermosipho africanus VEQIKAGNDLIMPGKTYNVFKDRKDEIKELKQAYEKGEITDDIINERVRT 296 Fervidobacterium nodosum AKQMAAGNDLIMPGKSHQVLKHRRNEIDDIRKAIENGELTEEVLNERIRN 299 Thermotoga maritima VEQLKAGNDMIMPGKAYQVNTERRDEIEEIMEALKEGKLSEEVLDECVRN 299 Thermotoga neapolitana VEQLKAGNDLIMPGKAYQVNTERRDEIEEIMEALKEGKLSEEVLDECVRN 299 Thermofilum sp. VRQINAGNDVIMPG--------GQDKLEQVLKAVREGKIPIETIDRAVTR 280 Thermofilum pendens VEQVNAGNDLIMPG--------SDEAVEKLLEAARSGRLRLEALEASAER 268 Caldivirga maquilingensis IEQVKAGNDLIMPG--------SDEIVSRLIDAVKRGELSEDYVNRSAAR 273 Thermotoga naphthophila ILKVLVNAPSFKGYRYSNKPDLESHAEVAYKAGAEGVVLLENNGVLPFDE 349 Thermotoga petrophila ILKVLVNAPSFKGYRYSNKPDLESHAKVAYEAGVEGVVLLENNGVLPFDE 350 Thermosipho africanus ILNILMKTPSFKGYKYSNAPDFEKHARISYNAASEGVVLLKNNDVLPFSA 346 Fervidobacterium nodosum ILRVLVKMPSFKGYNYSNNPDLEKNAKISYEAGCEGVVLLKNNEVLPISK 349 Thermotoga maritima ILKVLVNAPSFKGYRYSNKPDLESHAEVAYEAGAEGVVLLENNGVLPFDE 349 Thermotoga neapolitana ILKVLVNAPSFKNYRYSNKPDLEKHAKVAYEAGAEGVVLLRNEEALPLSE 349 Thermofilum sp. VLSKLLGSAGYK--STPGKPDLEAHAKIAYEAAVEGMVLLKNEGALPIDR 328 Thermofilum pendens VLRLVRKSLTYRGYRPRGAPDLDGHARVAYEAASEGVVLLKNEGALPLGP 318 Caldivirga maquilingensis VLEFIKRTLAYKGYKPTNSPNLKEHAKLAYEAAAEGVILLKNNDALPLNA 323

Page 149: A THESIS TITLED - prr.hec.gov.pk

Results

119

Thermotoga naphthophila NTHVAVFGTGQIETIKGGTGSGDTHPRYTISILEGIKERNMKFDEELAST 399 Thermotoga petrophila SIHVAVFGTGQIETIKGGTGSGDTHPRYTISILEGIKERNMKFDEELTSI 400 Thermosipho africanus DAKVSIFGTGQIETVKGGFGSGDTHPRYTISIFEGFKEKGVKVDEKIGNF 396 Fervidobacterium nodosum DTTIALFGTGQIETIRGGTGSGETHPMYTINFLDGVVERGLNFDKELAQF 399 Thermotoga maritima NTHVAVFGTGQIETIKGGTGSGDTHPRYTISILEGIKERNMKFDEELAST 399 Thermotoga neapolitana NSKIALFGTGQIETIKGGTGSGDTHPRYAISILEGIKERGLNFDEELAKT 399 Thermofilum sp. GKKIAFFGVGQILTVKGGMGSGHTHPPYVSTILDAARERSLAIDEELAKK 378 Thermofilum pendens GARVALFGTGQVETLKGGMGSGHTHPRYVVTVLEGLKSGGLLVDEELSSI 368 Caldivirga maquilingensis NARIALFGTGQVETNRGGLGSGHTHPRYFINILDGLRSRGLRIDEELSSI 373 Thermotoga naphthophila YEEYIKKMRETEEYKPRTDSWGTAIKPKLPENFLPEKEIKKAAKKNDVAV 449 Thermotoga petrophila YEDYIKKMRETEEYKPRTDSWGTVIKPKLPENFLSEKEIKKAAKKNDAAV 450 Thermosipho africanus YKEKVFELRNGNYRPNYVNEFNLKIPPKLPEDILDESMIDEASETNDLAI 446 Fervidobacterium nodosum YRAKIEELRNGEYRITRG-QWNEEIKPKLPENLFIVSQLEDIAKRNDVAI 448 Thermotoga maritima YEEYIKKMRETEEYKPRTDSWGTVIKPKLPENFLSEKEIKKAAKKNDVAV 449 Thermotoga neapolitana YEDYIKKMRETEEYKPRRDSWGTIIKPKLPENFLSEKEIHKLAKKNDVAV 449 Thermofilum sp. YAEAVSTYKEELEIFYRD----EPDKPSISEDVVGEADIYATAQRNDLAV 424 Thermofilum pendens YERYVREARGEEFLEKLY--LDEVYADPLPQDIVSEGDAARFAERNDAAV 416 Caldivirga maquilingensis YVNYVKENRGEDYLCALY--YEEAYSEPLPQDIISEEQVRKYAERNDAAI 421

* Thermotoga naphthophila VVISRISGEGYDRKPVKGDFYLSDDELELIKTVSKEFHDQGKKVVVLLNI 499 Thermotoga petrophila VVISRISGEGYDRKPVKGDFYLSDDELELIKTVSREFHEQGKKVVVLLNI 500 Thermosipho africanus IVISRISGEFVDRRAVKGDYYLSDDEQKLIENVSKKFHSKGKKVIVLLNI 496 Fervidobacterium nodosum IFITRISGEGYDRRAEKGDYYLTDDEYELIKNVSETFHKYGKKSLVVLNI 498 Thermotoga maritima VVISRISGEGYDRKPVKGDFYLSDDELELIKTVSKEFHDQGKKVVVLLNI 499 Thermotoga neapolitana IVISRISGEGYDRKPVKGDFYLSDDETDLIKTVSREFHEQGKKVIVLLNI 499 Thermofilum sp. IVVTRVSGEGWDLAP--EDFYLRDDEKWLIDTVSEAYRKIGKPVVAILNI 472 Thermofilum pendens VVLYRVSGEGWDRRPVRGDFYLTESEERLLRLVSREFRGRGKKVVVVLNV 466 Caldivirga maquilingensis VVISRNSGEGWDRRAVKGDYYLTDSERRLIEIVSRQFHALGKKVTVLLNI 471 Thermotoga naphthophila GSPIEVASWRDLVDGILLVWQAGQEMGRIVADVLVGKINPSGKLPTTFPK 549 Thermotoga petrophila GSPIEVASWRDLVDGILLVWQAGQEMGRIVADVLVGRVNPSGKLPTTFPK 550 Thermosipho africanus GGPIEIASWIELVDGLLLIWQPGQEAGRVVADVCLGTVNPSGKLPTTFPK 546 Fervidobacterium nodosum GSPIEVESWKELTDGILLVWQPGQEAGRIFADVISGKVNPSGKLATTFPK 548 Thermotoga maritima GSPIEVASWRDLVDGILLVWQAGQEMGRIVADVLVGKINPSGKLPTTFPK 549 Thermotoga neapolitana GSPVEVVSWRDLVDGILLVWQAGQETGRIVADVLTGRINPSGKLPTTFPR 549 Thermofilum sp. GTPIDVASWREKVDAILLAWQPGQEAGRAIIDTLLGLVNPSGKLPTTFPK 522 Thermofilum pendens CGPIEVASWRDLVDAILVVWLPGQEAGRVVADVLAGRVNPSGKLPMTWPR 516 Caldivirga maquilingensis PAPIEVASWRDLVDAILLVWLPGQEAGRVIADALIGVVNPSGKLPVTFPK 521 Thermotoga naphthophila DYSDVPSWT----FPGEPKDNPQRVVYEEDIYVGYRYYDTFGVEPAYEFG 595 Thermotoga petrophila DYSDVPSWT----FPGEPKDNPQRVVYEEDIYVGYRYYDTFGVEPAYEFG 596 Thermosipho africanus DYQDIPSKS----FPGKPVENPLEVVYEEDIYVGYRYYDTFQIDPLFEFG 592 Fervidobacterium nodosum DYKDVPSRS----FPGEPKENPTSVTYDEGIYVGYRYYDTFRVEPSFEFG 594 Thermotoga maritima DYSDVPSWT----FPGEPKDNPQRVVYEEDIYVGYRYYDTFGVEPAYEFG 595 Thermotoga neapolitana DYSDVPSWT----FPGEPKDNPQKVVYEEDIYVGYRYYDTFGVEPAYEFG 595 Thermofilum sp. TLGDVPSWT----FGGEPAGSPKSVTYEEDIYVGYRYYDTFGKEPAYEFG 568 Thermofilum pendens DWTDVPAAKAPECYPGLPVEDPRRVVYCEGVYVGYRYYDTFGVEPAYEFG 566 Caldivirga maquilingensis DWGDVPTSKSPECYPGIPKENPGAVRYCEGIYVGYRYYDKYSVEPAYEFG 571 Thermotoga naphthophila YGLSYTKFEYKDLKIAIDGDILRVSYTITNTGDRAGKEVSQVYVKAPKGK 645 Thermotoga petrophila YGLSYTKFEYKDLKIAIDGDILRVSYTITNTGDRAGKEVSQVYVKAPKGK 646 Thermosipho africanus YGLSYTKFDYKNLIVKHDDENIEICFEIENSGNVEGKEIAQVYVKAPKAR 642 Fervidobacterium nodosum FGLSYTKFEYSNLNLYKDGSSIVVCFDVKNVGNVPGKEITQLYVRAPRVK 644 Thermotoga maritima YGLSYTKFEYKDLKIAIDGETLRVSYTITNTGDRAGKEVSQVYIKAPKGK 645 Thermotoga neapolitana YGLSYTTFEYSDLNVSFDGETLRVQYRIENTGGRAGKEVSQVYIKAPKGK 645 Thermofilum sp. YGLSYTTFSYSDLSIEQDSEQVKVKFRVTNTGKMPGKEVAQVYVKPPKGK 618 Thermofilum pendens YGLSYTKFEYRGLRVALSRGALKVSFEVVNAGSRPGKEVAQVYVRAPRGR 616 Caldivirga maquilingensis YGLSYTRFEYRGLSVVKDGELIKVSFDVVNVGKHPGKEVAQVYVKAPQGK 621

Page 150: A THESIS TITLED - prr.hec.gov.pk

Results

120

Thermotoga naphthophila IDKPFQELKAFHKTKLLNPGESEKIFLEIPLRDLASFDGKE--WVVESGE 693 Thermotoga petrophila IDKPFQELKAFHKTKLLNPGESEKIFLEIPLRDLASFDGKE--WVVESGE 694 Thermosipho africanus LDKPFQELKGFYKTKMLKPGEKEEISIKIALRDLTSFCKDK--WVLEEGE 690 Fervidobacterium nodosum IDKPYQELKGFFKTDTLAPGEMQKVKITVQISDLASFDGEK--WVVEEGI 692 Thermotoga maritima IDKPFQELKAFHKTKLLNPGESEEISLEIPLRDLASFDGKE--WVVESGE 693 Thermotoga neapolitana IDKPFQELKAFHKTRLLNPGESEEVVLEIPVRDLASFNGEE--WVVEAGE 693 Thermofilum sp. IDKPQKELKAFKKTRLLQPGEHEDIVLTIGYKDMASFNGKE--WVLEKGT 666 Thermofilum pendens IDKPFQELKAFRKTRLLEPGEAERIKLRVSLRDLASFDEREKVWVVEPGE 666 Caldivirga maquilingensis LDKPVQELKAFKKTRLLNPGELEHVELTINVRDLASFDESRGMWIIDDGE 671 Thermotoga naphthophila YEVRVGASSRDIRLKDVFLVEEEKRFKP 721 Thermotoga petrophila YEVRVGASSRDIRLRDIFLVEGEKRFKP 722 Thermosipho africanus YRIRVGASSRDIRLEEKVFLKE------ 712 Fervidobacterium nodosum YEFRVGASSRDIRLK------------- 707 Thermotoga maritima YEVRVGASSRDIRLRDIFLVEGEKRFKP 721 Thermotoga neapolitana YEVRVGASSRNIKLKGTFSVGEERRFKP 721 Thermofilum sp. YKIMVGSSSRRIHLEGEFKIHEEKTFR- 693 Thermofilum pendens YEVRVGSSSRDIRLTEHFEVKQELRFAP 694 Caldivirga maquilingensis YEVRVGASSRDIRLTGKFTVSGVIEFKP 699

Figure 4.11 (b): Multi-alignment of β-glucosidase from T. naphthophila RKU-10T (TnBglB) with some

other GH3 family β-glucosidases. Highly conserved amino acid residues are indicated by blocks. The

catalytic sites are represented by asterisk (*) on the top of alignment, the conserved catalytic Aspartic acid

residue act as nucleophile active site (D242) and conserved Glutamic acid act as catalytic proton donor (E458)

and. Thermotoga naphthophila (ADA66752.1), Thermosipho africanus (WP_012580493.1),

Fervidobacterium nodosum (WP_011994742.1), Thermotoga maritima (WP_004082478.1), Thermotoga

petrophila (ABQ46915.1), Thermotoga neapolitana (WP_015919165.1), Thermofilum sp.

(WP_020961743.1), Thermofilum pendens (WP_011753156.1), Caldivirga maquilingensis

(WP_012186357.1).

4.4.17.1. TnBglA Prediction of Signal Peptide

By using online SignalP4.1 prediction server (predicts the presence and location of signal

peptide sequences), determined the presence of signal peptide sequence in the TnBglA

protein. After studied the translation sequence, it was confirmed that TnBglA protein has

no putative signal peptide sequence (Figure 4.12a). SignalP 4.1 neutral network produced

three scores in the form of output for each regions (position) in the sequence of TnBglA

amino acid input. In result, C-score (raw cleavage site score) represented the signal peptide

cleavage sites (immediately after the cleavage site showed high at the position). Signal

peptide score (S-score), output from the SignalP networks are used to discriminate

positions within signal peptides from mature part (position) of the proteins and from

without signal peptides proteins. Whereas, combined cleavage site score (Y-score) is an

average of the slope of S-score and C-score which predicted the cleavage site in a better

Page 151: A THESIS TITLED - prr.hec.gov.pk

Results

121

way as compare to the raw C-score alone. The maximum C and S-score for position 24

were 0.141 and 0.148, respectively (Table 4.5a). The SignalP results demonstrated that

there was no signal peptide probability.

Figure 4.12 (a): Signal peptide prediction graph of TnBglA

Table 4.5 (a): Output data of SignalP 4.1 for TnBglA Signal peptide prediction.

Measure Position Value Cutoff signal peptide

Max. C-score 24 0.141

0.450 NO

Max. Y-score 24 0.148

Max. S-score 10 0.217

Mean S 1-23 0.157

D-score 1-23 0.153

C-score= Raw cleavage site score, Y-score= Combined cleavage site score, S-score= Signal peptide

score, D= Discrimination.

4.4.17.2. TnBglB Prediction of Signal Peptide

The amino acid sequence of TnBglB was analyzed with the help of online SignalP4.1 server

and determined the presence or absence of signal peptide sequence. After using SignalP4.1

prediction server, it was confirmed that TnBglB protein has no putative signal peptide

Page 152: A THESIS TITLED - prr.hec.gov.pk

Results

122

sequence (Figure 4.12b). The maximum C-score, Y-score and S-score were 0.122 for

position 31, 0.109 for position 49 and 0.146 for position 36, respectively. All output results

of TnBglB for signal peptide prediction was mention in the following table 4.5b.

Figure 4.12 (b): Signal peptide prediction graph of TnBglB

Table 4.5 (b): Output data of SignalP 4.1 for TnBglB Signal peptide prediction.

Measure Position Value Cutoff Signal Peptide

(SP)

Max. C-score 31 0.122

0.450 NO

Max. Y-score 49 0.109

Max. S-score 36 0.146

Mean S 1–48 0.099

D-score 1–48 0.104

C-score= Raw cleavage site score, Y-score= Combined cleavage site score, S-score= Signal peptide

score, D= Discrimination.

Page 153: A THESIS TITLED - prr.hec.gov.pk

Results

123

4.4.18. Theoretical Properties of TnBglA and TnBglB Proteins

Many Physical and chemical properties of a given protein sequence was computed by using

ProtParam (ExPASy–Bioinformatics) tool. The amino acid sequences of TnBglA and

TnBglB were submitted to compute all parameters including amino acid composition,

aliphatic index, instability index, extinction coefficient, theoretical pI value, atomic

composition and GRAVY, which are listed in the table 4.6. According to the study of

ProtParam tool, the instability index of TnBglA and TnBglB are computed to be 31.23 and

30.83, respectively which classifies both proteins as stable.

Table 4.6: Theoretical Properties of TnBglA and TnBglB Protein using ProtParam tool.

Properties TnBglA TnBglB

Number of amino acids 446 721

Molecular weight 51.509 kDa 81.142 kDa

Positively charged residues (Arg+Lys) 51 94

Negatively charged residues (Asp+Glu) 63 116

Theoretical pI 5.56 5.19

Extinction coefficients (M-1 cm-1) 121240 102930

Instability index 31.23 30.83

Aliphatic index 82.60 84.17

Formula C2376H3537N611O664S7 C3648H5702N958O1099S18

GRAVY -0.373 -0.427

Total number of atoms 7195 11425

GRAVY = Grand average of hydropathicity, Asp = Aspartic acid, Arg = Arginine, Lys = Lysine,

and Glu = Glutamic acid.

Page 154: A THESIS TITLED - prr.hec.gov.pk

Results

124

4.5. Sub-cloning of TnbglA and TnbglB in Expression Vector

To regulate the adequate level of genes expression, an excellent vector pET-21a(+) was

selected for the construction of recombinant expression plasmids. TnbglA and TnbglB

genes were sub-cloned in pET-21a(+) vector and overexpressed under the control of T7/lac

promoter system in host cells. Following are the steps of sub-cloning of genes in expression

plasmids.

4.5.1. Double Restriction of Recombinant Plasmids

Recombinant pTZ57R/T–TnbglA and pTZ57R/T–TnbglB plasmids were double restricted

with NdeI and HindIII in separate individual sterile tubes followed by electrophoresed on

0.8% agarose gel and evaluated the resultant fragment bands under UV-light. Double

cleaved recombinant pTZ57R/T–TnbglA plasmids from selected positive transformants

gave three prominent bands of ~4.227 kb, 2.886 kb and 1.341 kb bands on gel that

represented the uncut recombinant plasmid, pTZ57R/T vector and lastly insert TnbglA

fragment, respectively (Figure 4.13a).

Figure 4.13 (a): Analysis of recombinant pTZ57R/T–TnbglA plasmid after double digestion using two

restriction endonucleases. Lane M, DNA ladder; Lane 1-8, restricted plasmids with NdeI and HindIII and

three resultant bands of approximately 4.227 kb (uncut plasmid), 2.886 kb (pTZ57R/T) and 1.341 kb (insert

TnbglA gene) were observed.

kb

~ 4.227 kb

2.886 kb

1.341 kb

0.5

8.0

5.0

3.0

1.0

1.5

M 1 2 3 4 5 6 7 8

Page 155: A THESIS TITLED - prr.hec.gov.pk

Results

125

After double digestion of recombinant plasmids pTZ57R/T–TnbglB isolated from selected

positive transformants colonies, three bands of ~5.052 kb (uncut recombinant plasmid),

2.886 kb (linerized pTZ57R/T) and 2.166 kb (insert TnbglB) bands were observed on

agarose gel along with DNA ladder (Figure 4.13b).

Figure 4.13 (b): Analysis of recombinant pTZ57R/T–TnbglB plasmid after double digestion using two

restriction endonucleases. Lane M, DNA size ladder; Lane 1-6, double restricted plasmids with NdeI and

HindIII enzymes, and three resultant bands of approximately 5.052 kb (uncut plasmid), 2.886 kb (pTZ57R/T)

and 2.166 kb (insert TnbglB gene) were observed.

4.5.2. Isolation and Double Restriction of pET-21a(+)

Glycerol stock of E. coli DH5α harboring expression vector pET-21a(+) was used to streak

freshly prepared LB-ampicillin plate followed by kept for overnight at 37°C. Next day, a

well isolated single colony selected (Figure 4.14) and inoculated in fresh sterile LB-

ampicillin broth for mini-preparation. After plasmid pET-21a(+) extraction, treated with

RNase to purify the pET from RNA remains (Figure 4.15a). After that purified pET-21a(+)

was subjected to restriction with NdeI and HindIII subsequently analyzed on 0.8% agarose

gel, a prominent band of 5.443 kb was obtained (Figure 4.15b).

M 1 2 3 4 5 6

2.886 kb

kb

10

5.0

3.0

2.0 2.166 kb

1.0

~5.052 kb

Page 156: A THESIS TITLED - prr.hec.gov.pk

Results

126

Figure 4.14: Streak plate of E. coli DH5α harboring pET-21a(+)

Figure 4.15 (a): Isolation of plasmid pET-21a(+), Lane M, DNA size marker; Lane 1 and 2, uncut plasmid

treated with RNase. (b) Double restricted pET-21a(+) using NdeI and HindIII restriction endonucleases, M,

DNA marker; Lane 1-4, bands of 5.443 kb linearized pET-21a(+) after double digestion.

4.5.3. Purification of Double Digested pET–21a(+), TnbglA and TnbglB

Double restricted fragments of pET-21a (5.443 kb), TnbglA (1.341 kb) and TnbglB (2.166

kb) were excised from gel using extraction kit. To evaluate the purity of gene clean double

digested DNA fragments, a small amount (2 µL) of purified products were loaded in the

wells of 0.8% agarose gel and purified products of double digested genes (TnbglA and

TnbglB) fragments and pET were observed under UV-light after electrophoresed (Figure

4.16a and 4.16b).

M 1 2 3 4 M 1 2

Uncut

pET-21a kb

3.0

1.0

0.5

5.443 kb

kb

4.0

5.0

3.0

1.0

0.5

(a) (b)

Page 157: A THESIS TITLED - prr.hec.gov.pk

Results

127

Figure 4.16 (a): Purified double restricted pET-21a(+), Lane M, DNA size marker; Lane 1 and 2, linearized

double cleaved pET-21a(+) after gene clean (5.443 kb). (b) Purified double digested DNA fragments (TnbglA

and TnbglB) after gene clean. Lane M, DNA marker; Lane 1 and 2, purified TnbglB (2.166 kb); Lane 3 and

4 purified TnbglA (1.341 kb).

4.5.4. Ligation and Transformation of E. coli DH5α with Constructed Plasmids

Purified double digested expression vector pET-21a(+) and genes fragments (TnbglA and

TnbglB) were ligated together using T4 DNA ligase followed by incubated at 16°C for 16-

18 hours. Ligation products of both genes (TnbglA and TnbglB) were used to transformed

competent cells of strain DH5α followed by spread transformants of both genes separately

on freshly prepared LB-agar plates containing ampicillin (100 µg mL-1) and kept for

overnight at 37°C incubator. Many white colonies were appeared on both (TnbglA and

TnbglB) LB-agar plates (Figure 4.17). Positive putative transformants were screened by

colony PCR and confirmed by double restriction analysis.

4.5.5. Colony PCR

Selected well isolated white colonies from both plates (TnbglA and TnbglB) and analyzed

the presence of insert gene in pET-21a(+) by colony PCR. After PCR process, all reactions

were loaded on 0.8% agarose gel separately and screened out the positive recombinant

colonies for both genes (Figure 4.18a and 4.18b).

M 1 2 M 1 2 M 3 4

(a) (b)

kb

8.0

kb

2.166 kb

1.341 kb 2.0

10 8.0 6.0 5.0 4.03.0 2.0 1.0 0.5 0.4 0.3 0.2

3.0

5.443 kb

1.2

0.5

Page 158: A THESIS TITLED - prr.hec.gov.pk

Results

128

Figure 4.17: Several white bacterial colonies were appeared on LB-agar plates supplement with ampicillin

after transformation of E. coli DH5α with constructed expression plasmids. (a) Control plate has shown

transformants colonies harboring circular pET-21a(+). (b) Positive putative transformants harboring

recombinant plasmids pET-21a–TnbglA. (c) Positive putative transformants colonies harboring recombinant

plasmids pET-21a–TnbglB.

Figure 4.18: Colony PCR for the screening of positive transformants of E. coli DH5α harboring constructed

recombinant plasmids. (a) Screening of positive transformants harboring pET-21a–TnbglA plasmids by

colony-PCR. Lane M, DNA marker; Lane 1-7, represent 1.341 kb TnbglA amplicons of different colony-

PCR tubes and all selected colonies are positive. (b) Lane M, DNA ladder; Lane 1-6, show 2.166 kb TnbglB

amplicons of different selected colony-PCR and all colonies are positive.

(a) Control Plate (b) pET-21a–TnbglA Plate (c) pET-21a–TnbglB Plate

M 1 2 3 4 5 6 7

(a) (b)

M 1 2 3 4 5 6

10 8.0 6.0 5.0 4.03.0 2.0 1.5 1.00.80.5 0.4 0.3 0.2

kb

1.34 kb

2.166 kb

Page 159: A THESIS TITLED - prr.hec.gov.pk

Results

129

4.5.6. Extraction of Recombinant Expression Plasmids

After colony PCR, positive bacterial transformants harboring recombinant expression

plasmids (pET-21a–TnbglA and pET-21a–TnbglB) were further confirmed by double

restriction analysis. Therefore, recombinant plasmids were isolated from particular positive

cloned E. coli bacterial colonies, and extracted plasmids after RNase treatment were

visualized and analyzed by agarose gel electrophoresis (Figure 4.19a and 4.19b).

Figure 4.19: Isolated recombinant plasmids treated with RNase (a) M, DNA marker; Lane 1-3, uncut circular

recombinant pET-21a–TnbglA plasmids. (b) M, DNA marker; Lane 1-4, uncut circular recombinant pET-

21a–TnbglB plasmids from selected positive transformants.

4.5.7. Restriction Analysis of Recombinant Plasmids

To confirm the positive transformants, double digestion analysis of isolated recombinant

plasmids pET-21a–TnbglA and pET-21a–TnbglB were carried out in sterile individual

tubes with NdeI and HindIII restriction endonucleases followed by analyzed on 0.8%

agarose gel electrophoresis (Figure 4.20a and 4.20b).

4.5.8. Transformation of E. coli BL21 CodonPlus (DE3) with Constructed Expression

Plasmids

After the confirmation of insert genes by restriction analysis, the isolated recombinant

plasmids (pET-21a–TnbglA and pET-21a–TnbglB) were used to transform E. coli

CodonPlus competent cells as a mesophilic expression host. Bacterial transformed spread

10 8.0 6.0 5.0 4.03.0 2.0 1.5 1.00.70.5 0.4 0.3 0.2 0.1

kb

1 2 3 M M 1 2 3 4

(a) (b)

Uncut

Plasmid

Page 160: A THESIS TITLED - prr.hec.gov.pk

Results

130

on LB-agar plates containing ampicillin and chloramphenicol (100 µg mL-1 and 50 µg mL-

1, respectively) followed by incubated for overnight at 37°C, a number of white colonies

obtained on plates (Figure 4.21a and 4.21b).

Figure 4.20: Analysis of recombinant constructed expression plasmids after double digestion using

restriction endonucleases. (a) Lane M, DNA ladder; Lane 1-4, double restricted pET-21a–TnbglA plasmids

with NdeI and HindIII enzymes and resultant three bands of ~ 6.784 kb (uncut plasmid), 5.443 kb (linearized

pET-21a) and 1.341 kb (insert TnbglA gene) were observed. (b) Lane M, DNA size ladder; Lane 1-4, double

restricted pET-21a–TnbglB plasmids with NdeI and HindIII, and three bands were observed, in which first

one was ~7.609 kb (uncut plasmid), second 5.443 kb (pET-21a) and third 2.166 kb (insert TnbglB gene)

fragments.

Figure 4.21: Several white bacterial colonies were appeared on LB-agar plates supplement with ampicillin

and chloramphenicol after transformation of E. coli BL21 CodonPlus (DE3)-RIPL with recombinant circular

plasmids. (a) Plate represent transformants colonies harboring recombinant plasmids pET-21a–TnbglA (b)

Transformants colonies harboring recombinant plasmids pET-21a–TnbglB.

10 8.0 6.0 5.0 4.03.0 2.0 1.5 1.00.5

~6.784 kb ~7.609 kb

5.443 kb

2.166 kb

5.443 kb

1.341 kb

(a) (b)

M 1 2 3 4 M 1 2 3 4 kb

(a) pET-21a–TnbglA in BL21 (b) pET-21a–TnbglB in BL21

Page 161: A THESIS TITLED - prr.hec.gov.pk

Results

131

4.5.9. Colony PCR

To screen the positive transformed colonies of pET-21a–TnbglA and pET-21a–TnbglB,

colony PCR was performed individually using specific primers at optimal standard

conditions of PCR as described earlier. The result of colony PCR of both recombinant

colonies were analyzed after electrophoresis and observed that all selected colonies were

positive for both cloned genes (Figure 4.22a and 4.22b).

Figure 4.22: Colony PCR for the screening of positive transformants of E. coli BL21 CodonPlus harboring

constructed recombinant plasmids. (a) Transformants harboring pET-21a–TnbglA plasmids confirmed by

colony-PCR. Lane M, DNA marker; Lane 1-6, represent 1.341 kb TnbglA amplicons after colony-PCR and

all randomly selected colonies are positive. (b) Lane M, DNA ladder; Lane 1-6, show 2.166 kb TnbglB

amplicons after colony-PCR and all selected colonies are positive.

4.6. Expression Analysis of Recombinant Proteins (TnBglA and TnBglB)

The positive bacterial transformants of E. coli CodonPlus harboring pET-21a–TnbglA and

pET-21a–TnbglB plasmids were grown separately in LB-broth medium supplemented with

ampicillin and chloramphenicol (100 µg mL−1 and 50 µg mL−1, respectively), induced

initially with 0.5 mM IPTG and kept in a shaking incubator (200 rev min-1) at 37°C for the

production of recombinant enzymes. Initially, heterologous crude proteins (TnbglA and

TnbglB) expression in supernatant were analyzed after 24 h by 12% SDS-PAGE along with

2.166

kb

kb kb

1.341

kb

10 8.0 6.0 5.0 4.03.5 3.0 2.5 2.0 1.51.2 1.0 0.9 0.8 0.6 0.5

M 1 2 3 4 5 6 M 1 2 3 4 5 6

(a) (b) 10 8.0 6.0 5.0 4.03.5 3.0 2.5 2.0 1.51.2 1.0 0.9 0.8 0.6 0.5

Page 162: A THESIS TITLED - prr.hec.gov.pk

Results

132

crude control supernatant of E. coli BL21 CodonPlus harboring pET-21a (+) with and

without induction (Figure 4.23a and 4.23b). Recombinant protein TnBglA and TnBglB

displayed a prominent band of 51.5 kDa and 81 kDa molecular weight, respectively on

SDS-PAGE; no such bands were observed in the control samples.

Figure 4.23: SDS-PAGE analysis of recombinant proteins expression in E. coli BL 21 CodonPlus DE3-

(RIPL). (a) Heterologous expression of TnBglA by 12% SDS-PAGE. Lane M, Protein marker (cat # 69079-

3); Lane 1, Crude control supernatant harboring pET-21a (+) after induction with 0.5 mM IPTG; Lane 2,

Crude control supernatant harboring pET-21a(+) without induction; Lane 3, Crude supernatant harboring

pET-21a–TnBglA after induction with 0.5 mM IPTG. (b) Heterologous expression of TnBglB. Lane M,

Protein marker; Lane 1, Crude control supernatant harboring pET-21a(+) after induction with 0.5 mM IPTG;

Lane 2, Crude control supernatant harboring pET-21a(+) without induction; Lane 3, Crude supernatant

harboring pET-21a–TnBglB after induction with 0.5 mM IPTG.

4.7. Preliminary Characterization of TnBglA and TnBglB

Initially, both cloned cells were cultivated in individual sterilized LB-medium, to evaluate

the preliminary characterization of both recombinant enzymes. IPTG (0.5 mM) used to

induce the cultures after that they kept for 96 hours at 37°C and were withdrawn the sample

at regular intervals. After centrifugation, the supernatant (extracellular), cell-free lysate

225 150

100

kDa

(a) (b)

51.5 kDa

81 kDa

35

25

15

10

50

75

25

35

50

75

100

150

225

kDa

Page 163: A THESIS TITLED - prr.hec.gov.pk

Results

133

(intracellular) and cell pellet (cell-bound) fractions of culture were analyzed by SDS-PAGE

and assayed to determine recombinant enzyme activity. Preliminary, optimal temperature

and pH were evaluated to perform activity assay for both recombinant enzymes.

4.7.1 Optimal buffer system for TnBglA and TnBglB

Optimal pH and most suitable buffer for the recombinant TnBglA and TnBglB enzymes

were evaluated by using various buffering system such as, McIlvaine buffer (for pH 2.2-

8.0), sodium acetate buffer (for pH 3.7-5.6), Tris-Cl buffer (for pH 7.0-9.0), MES buffer

(for pH 5.6-6.6), Sorensen’s phosphate buffer (for pH 5.8-8.0), HEPES buffer (for pH 6.8-

8.2), MOP buffer (for pH 7.0-7.8), and CAPS buffer (for pH 9.0-11.0). pH profile was

determined at 70°C for 10 min incubation against pNPG (4 mg mL-1) as substrate, at

different pH values ranging from 3.0-11.0 using various buffer system. Low activity was

observed in MES buffer and MOP buffer. Finally, McIlvaine buffer system was found

optimal buffering system for both cloned enzymes and maximum activity was found at pH

7.0 and pH 5.0 for crude TnBglA and TnBglB, respectively (Figure 4.24a and 4.24b).

4.7.2. Optimal Temperature for TnBglA and TnBglB

The optimal temperature of cloned TnBglA and TnBglB were measured over a broad

temperature ranged from 30-100°C (increments of 5°C), using pNPG (4 mg mL-1) as

substrate, in optimal pH buffer system for both enzymes that was McIlvaine buffer having

pH 7.0 for TnBglA and pH 5.0 for TnBglB. Maximum TnBglA and TnBglB activities were

observed at 95°C and 85°C temperature, respectively (Figure 4.25). All results related to

temperature profile was expressed as percentage of relative activity.

4.7.3. Incubation Time for Enzyme Assay

To find out the optimal incubation time for the hydrolysis of pNPG (4 mg mL-1) substrate,

activity assays were performed for both recombinant enzymes (TnBglA and TnBglB).

Enzyme-substrate reaction mixtures for TnBglA and TnBglB were incubated for 1–15

minutes at their respective optimal conditions, individually. Reactions were withdrawn

periodically and scrutinized the optimum time for the substrate hydrolysis. Maximum

Page 164: A THESIS TITLED - prr.hec.gov.pk

Results

134

activity of TnBglA and TnBglB were observed after 10 minutes incubation at 95°C and

85°C temperature, respectively (Figure 4.26).

Figure 4.24: Effect of pH and various buffer system on recombinant enzymes. Sodium acetate buffer (closed

diamond, pH 4.0-5.5), McIlvaine buffer (closed square, pH 5.0-7.5), Tris-Cl buffer (open triangle, pH 7.0-

9.0), HEPES buffer (closed circle, pH 7.0-8.0), phosphate buffer (opened circle, for pH 6.0-8.0), and CAPS

buffer (dash, pH 9.0-11.0) buffers. (a) The maximum TnBglA activity was observed at pH 7.0 with McIlvaine

buffer. (b) TnBglB showed maximum activity at pH 5.0 with McIlvaine buffer, both are defined as 100%

activity to calculate the relative activity. Reported data is the average of at least three independent

experiments with the standard deviation (+ SD) presented as error bars, which differ significantly at p≤0.05.

0

20

40

60

80

100

120

4 4.5 5 5.5 6 6.5 7 7.5 8 8.5 9 9.5 10 10.5 11

Rel

ati

ve

act

ivit

y %

pH

(a) Effect of various buffer on TnBglA activity

0

20

40

60

80

100

120

4 4.5 5 5.5 6 6.5 7 7.5 8 8.5 9 9.5 10 10.5 11

Rel

ativ

e ac

tivi

ty %

pH

(b) Effect of various buffer on TnBglB activity

Page 165: A THESIS TITLED - prr.hec.gov.pk

Results

135

Figure 4.25: Effect of temperature on TnBglA and TnBglB. TnBglA displayed optimal activity at 95°C while

TnBglB showed optimal catalytic activity at 85°C, both are defined as 100% activity to calculate the relative

activity. Y-error bars show the standard deviation (±SD) of parallel three experimental reactions. Probability

of all the variables is < 5% significance level, which entail that elected variables are significant at 95%

confidence level.

Figure 4.26: Effect of time course profile of recombinant enzymes. TnBglA and TnBglB displayed optimal

catalytic activity after 10 minutes of incubation at 95°C and 85°C, respectively. Both are defined as 100%

activity to calculate the relative activity. Reported values in results are the average of three independent

experiments, Y-error bars show the standard deviation (±SD) among three reactions, which differ

significantly at p≤0.05.

0

20

40

60

80

100

120

45 50 55 60 65 70 75 80 85 90 95 100

Rel

ati

ve

act

ivit

y %

Temperature oC

Effect of Temperature on TnBglA and TnBglB activity

TnBglA TnBglB

0

20

40

60

80

100

120

0 5 10 15

Rel

ativ

e ac

tivi

ty %

Time (minute)

Effect of Incubation Time for Activity Assay

TnBglA TnBglB

Page 166: A THESIS TITLED - prr.hec.gov.pk

Results

136

4.7.4. Cell fractionation

Heterologous expression and activity of proteins (TnBglA and TnBglB) were analyzed in

all three fractions: extracellular, intracellular and cell-bound fractions. All fractions have

exhibited expression and enzymes activity while highest TnBglA and TnBglB activities

were observed in soluble extracellular fraction (culture supernatant) followed by the

intracellular and the cell-bound fractions (Figure 4.27a and 4.27b). For both enzymes,

maximum protein concentration was found in cell lysate fraction but optimal enzyme

activity was observed in culture supernatant (soluble extracellular fraction). As analysis

was conducted by SDS-PAGE, less expression of recombinant TnBglA and TnBglB

observed in cell lysate as compared to soluble fraction supernatant (Figure 4.28a and

4.28b).

Figure 4.27: Cell fractionation analysis for cloned TnBglA and TnBglB. (a) TnBglA represented optimal

activity (at 95°C using pH 7.0 buffer) and expression in soluble extracellular fraction. (b) TnBglB showed

peak catalytic activity (at 85°C, pH 5.0 buffer) and expression in soluble extracellular fraction, both are

defined as 100% activity to calculate the relative activity. All values shown in results are the average of

triplicate experiments. Y-error bars show the standard deviation (±SD) among three reactions, and probability

of all the variables is < 5% significance level, which entail that elected variables are significant at 95%

confidence level.

0

1

2

3

4

0

20

40

60

80

100

120

Cell lysate Mediasupernatant

Cell bound

Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

acti

vit

y %

Fractions

(a) Cell fractionation analysis of TnBglA

Enzyme Activity Protein Conc.

0

1

2

3

4

0

20

40

60

80

100

120

Cell lysate Mediasupernatant

Cell bound

Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

act

ivit

y %

Fractions

(b) Cell fractionation analysis of TnBglB

Enzyme Activity Protein Conc.

Page 167: A THESIS TITLED - prr.hec.gov.pk

Results

137

Figure 4.28: SDS-PAGE analysis of cell lysate (intracellular) and supernatant (extracellular) fractions of

recombinant proteins expressed in E. coli BL 21 CodonPlus DE3-(RIPL). (a) Expression of Intra- and

extracellular fractions of TnBglA by 12% SDS-PAGE. Lane M, Protein molecular weight marker; Lane 1,

Crude control cell lysate of E. coli harboring pET-21a (+) after induction with 0.5 mM IPTG; Lane 2, Crude

cell lysate fraction of host after induction with 0.5 mM IPTG; Lane 3, Crude extracellular supernatant fraction

of TnBglA after induction with 0.5 mM IPTG. (b) Heterologous expression of TnBglB by applying both

fractions. Lane M, Protein marker; Lane 1, Crude control cell lysate harboring pET-21a (+) after induction

with 0.5 mM IPTG; Lane 2, Crude supernatant harboring pET-21a (+) after induction with 0.5 mM IPTG;

Lane 3, Crude supernatant harboring pET-21a–TnBglB after induction with 0.5 mM IPTG; Lane 4, Crude

cell lysate of host cells harboring pET-21a–TnBglB after induction with 0.5 mM IPTG.

4.8. Production of Recombinant TnBglA and TnBglB

Engineered E. coli CodonPlus cells harboring pET-21a–TnbglA and pET-21a–TnbglB

plasmids, were used for the production of cloned enzymes, in shake flasks. Various

cultivation conditions and parameters including pre-induction optical density or induction

stage (influence the yield and reproducibility of expressed proteins), medium pH, heat

shock treatment, agitation, inducer concentrations, inducement temperature and time, were

optimized generally in LB medium to achieve maximum yield of heterologous proteins,

prior to preparative-scale experiments.

81 kDa 150

225

25

35

50

75

kDa

51.5 kDa

(a) (b)

Page 168: A THESIS TITLED - prr.hec.gov.pk

Results

138

4.8.1. Pre-induction optimal cell density

Optimum culture density of the E. coli BL21 CodonPlus (DE3) containing recombinant

plasmids (pET-21a–TnbglA and pET-21a–TnbglB) at the time of induction was determined

by using LB-broth medium supplied with both antibiotics (with same concentration as

described earlier), in the individual flasks. Recombinant genes were induced with IPTG

(0.5 mM) at different optical density of culture (OD600nm) 0.5 to 0.9; the optimal enzyme

activity and expression was observed when both recombinant bacterial cultures were

induced at 0.6 optical density (OD600nm) of E. coli cells (Figure 4.29a and 4.29b).

Figure 4.29: Effect of Pre-induction optimal cell density of cloned bacterial on recombinant enzymes

expression (a) Represents the optimal TnBglA activity (at 95°C using pH 7.0 buffer) and expression at 0.6

OD600nm. (b) TnBglB showed optimal catalytic activity (at 85°C, pH 5.0 buffer) and expression at 0.6 OD600nm,

both are defined as 100% activity to calculate the relative activity. Y-error bars show the standard deviation

(±SD) of parallel three experimental reactions. Probability of all the variables is < 5% significance level,

which entail that elected variables are significant at 95% confidence level.

0

0.5

1

1.5

2

2.5

3

0

20

40

60

80

100

120

0.5 0.6 0.7 0.8 0.9

Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ativ

e ac

tivi

ty %

OD600 nm

(a) Optimization of Cell density on TnBglA

Enzyme Activity Protein Conc.

00.511.522.53

020406080

100120

0.4 0.5 0.6 0.7 0.8 0.9

Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

act

ivit

y %

OD600nm

(b) Optimization of Cell Density on TnBglB

Enzyme Activity Protein Conc.

Page 169: A THESIS TITLED - prr.hec.gov.pk

Results

139

4.8.2. Effect of Heat Shock

To investigate the effect of heat shock treatment, host cells were incubated at 42°C (1 h)

before and after induction with IPTG, simultaneously a flask was run parallel in which

culture did not treat with heat shock. Direct induction of recombinant cells at OD600nm 0.6,

resulted in comparatively lower enzymes activity and expression as compared to when

bacterial culture was given a heat shock in a shaking water bath (200 rev min-1) after that

culture was induced with IPTG (0.5 mM). Both enzymes activities and expression were

also lower when heat shock was given after induction (Figure 4.30a and 4.30b).

Figure 4.30: Effect of heat shock treatment on recombinant TnBglA and TnBglB expression. (a) Represents

optimal TnBglA activity when culture gave heat shock before induction (HSBI) at 0.6 OD600nm. (b) Optimal

TnBglB expression was obtained when culture gave heat shock before induction (HSBI) at 0.6 OD600nm, both

are defined as 100% activity to calculate the relative activity. Y-error bars show the standard deviation (±SD)

of parallel three reactions. Probability of all the variables is < 5% significance level, which entail that elected

variables are significant at 95% confidence level. WHSI represented, without heat shock induction and HSAI,

heat shock after induction.

0

0.5

1

1.5

2

2.5

3

0

20

40

60

80

100

120

WHSI HSBI HSAI Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ativ

e a

ctiv

ity

%

Heat Shock Treatment

(a) Effect of Heat Shock on TnBglA

Enzyme Activity Protein Conc.

00.511.522.53

020406080

100120

WHSI HSBI HSAI

Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

act

ivit

y %

Heat Shock Treatment

(b) Effect of Heat Shock on TnBglB

Enzyme Activity Protein Conc.

Page 170: A THESIS TITLED - prr.hec.gov.pk

Results

140

4.8.3. Optimal IPTG concentration

TnbglA and TnbglB genes ligated into pET-21a (+), propagated and expressed in host

under the control of T7 RNA polymerase promoter. Therefore, to over-express

heterologous protein, induced the culture cells with different concentration of IPTG (0.1-

0.9 mM), which revealed that optimal level of recombinant TnBglA and TnBglB activity,

expression, cell density and culture proteins were observed with 0.5 mM concentration,

beyond 0.5 mM, no increase in expression level was observed on SDS-PAGE analysis

(Figure 4.31a and 4.31b). Moreover, further increment in IPTG concentration (0.9 mM)

retarded the growth of cloned bacterial cells as well as lowered the TnBglA and TnBglB

activities (Figure 4.32a and 4.32b).

Figure 4.31: Analysis of different concentration of IPTG as inducer on TnBglA and TnBglB expression. (a)

Expression of extracellular fractions of TnBglA induced with different concentration of IPTG. Lane C, Crude

control supernatant of E. coli harboring pET-21a (+) without induction; Lane 1, Crude extracellular

supernatant of recombinant bacterial culture after induction with 0.2 mM IPTG; Lane 2, Crude supernatant

after induction with 0.3 mM IPTG; Lane 3, Crude supernatant after induction with 0.4 mM IPTG; Lane 4,

Crude supernatant after induction with 0.5 mM IPTG; Lane 5, Crude supernatant after induction with 0.6

mM IPTG. (b) Heterologous expression of TnBglB, induced with different concentration of IPTG. Lane M,

Protein marker; Lane 1, Crude control supernatant harboring pET-21a (+) without induction; Lane 2, Crude

supernatant after induction with 0.2 mM IPTG; Lane 3, Crude supernatant after induction with 0.3 mM IPTG;

Lane 4, Crude supernatant after induction with 0.4 mM IPTG; Lane 5, Crude supernatant after induction with

0.5 mM IPTG; Lane 6, Crude supernatant after induction with 0.6 mM IPTG.

(a) (b)

C 1 2 3 4 5

51.5 kDa

81 kDa

Page 171: A THESIS TITLED - prr.hec.gov.pk

Results

141

Figure 4.32: Effect of different concentration of IPTG on recombinant enzymes expression (a) Optimal

TnBglA activity and expression was observed at 0.5 mM IPTG. (b) TnBglB showed optimal activity and

expression at 0.5 mM IPTG, both are defined as 100% activity to calculate the relative activity. Y-error bars

show the standard deviation (±SD) of parallel three experimental reactions, which significant at probability

5%.

4.8.4. Optimal induction temperature

Immediately after heat shock and induction, the bacterial culture was incubated at 16-42°C

temperatures in a shaking incubator at 200 rev min-1. Samples were withdrawn periodically

0

0.5

1

1.5

2

2.5

3

0

20

40

60

80

100

120

0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9

Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

act

ivit

y %

IPTG Conconcentration (mM)

(a) Optimization of IPTG Conc. on TnBglA

Enzyme Activity Protein Conc.

0

0.5

1

1.5

2

2.5

3

3.5

0

20

40

60

80

100

120

0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9

Pro

tein

Con

c. (

mg

mL

-1)

Rel

ativ

e ac

tivi

ty %

IPTG Concentration (mM)

(b) Optimization of IPTG Conc. for TnBglB

Enzyme Activity Protein Conc.

Page 172: A THESIS TITLED - prr.hec.gov.pk

Results

142

from each incubated flask for the analysis of TnBglA and TnBglB enzyme expression and

total cell protein. The optimal incubation temperature for maximum cell growth and

expression in terms enzymes activities was observed at 22°C for both cloned enzymes

(Figure 4.33a and 4.33b).

Figure 4.33: Effect of different induction temperature on recombinant TnBglA and TnBglB expression (a)

Optimal TnBglA expression was found when culture was induced at 22°C with 0.5 mM IPTG induction. (b)

TnBglB showed optimal at 22°C with 0.5 mM IPTG induction, both are defined as 100% activity to calculate

the relative activity. Y-error bars display the standard deviation (±SD) of parallel three experimental

reactions, which significant at probability 5%.

0

0.5

1

1.5

2

2.5

3

0

20

40

60

80

100

120

16 22 26 30 37 40 42 Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

acti

vity

%

Temperature (oC)

(a) Optimization of Induction Temperature of TnBglA

Enzyme Activity Protein Conc.

0

0.5

1

1.5

2

2.5

3

0

20

40

60

80

100

120

16 22 26 30 37 40 42 Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

act

ivit

y %

Temperature (oC)

(b) Optimization of Induction Temperature of TnBglB

Enzyme Activity Protein Conc.

Page 173: A THESIS TITLED - prr.hec.gov.pk

Results

143

4.8.5. Optimal agitation speed

Incubated the both recombinant bacterial culture after induction with 0.5 mM IPTG at

different agitation speed (50, 100, 150, 200 and 250 rev min-1) at 22°C. Best host culture

production, TnBglA and TnBglB activity were determined at an agitation speed of 200 rev

min-1 (Figure 4.34a and 4.34b).

Figure 4.34: Effect of agitation speed on culture growth and recombinant enzymes expression (a) Optimal

TnBglA expression was found when culture was incubated at 200 rev min-1 and 22°C temperature with 0.5

mM IPTG induction. (b) TnBglB optimal expression was obtained when host culture was incubated at 200

rev min-1 and 22°C with 0.5 mM IPTG induction, both are defined as 100% activity to calculate the relative

activity. Y-error bars show the standard deviation (±SD) of parallel three experimental reactions. Probability

of all the variables is < 5% significance level which entail that elected variables are significant at 95%

confidence interval.

0

0.5

1

1.5

2

2.5

3

0

20

40

60

80

100

120

50 100 150 200 250 Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

act

ivit

y %

Revolution minute-1

(a) Effect of Agitation on TnBglA Expression

Enzyme Activity Protein Conc.

0

0.5

1

1.5

2

2.5

3

3.5

0

20

40

60

80

100

120

50 100 150 200 250 Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ativ

e ac

tivi

ty %

Revolution minute-1

(b) Effect of Agitation on TnBglB Expression

Enzyme Activity Protein Conc.

Page 174: A THESIS TITLED - prr.hec.gov.pk

Results

144

4.8.6. Optimal Medium pH

To determine the optimum pH of medium for TnBglA and TnBglB production, proper

protein folding and for better growth of engineered bacterial cultures. Recombinant E. coli

culture was inoculated individually in different pH media ranging from 5.5-8.0 and

incubated at optimal conditions after induction with IPTG (0.5 mM). The highest yield of

both recombinant proteins TnBglA and TnBglB were observed at pH 7.0 (Figure 4.35a and

4.35b). Between pH 6.0-7.5, growth rate of cloned bacterial cells and 0.6 optimal density

(OD600nm) remained unaffected, while the heterologous protein secretion and overall

production of total cell protein (TCP) were affected remarkably, most probably due to

protease activity in fermentation culture broth. Bacterial transformants, therefore, grown

well in neutral pH medium at 22°C induction temperature.

00.511.522.533.544.5

0

20

40

60

80

100

120

5 5.5 6 6.5 7 7.5 8P

rote

in C

on

c. (

mg

mL

-1)

Rel

ati

ve

acti

vit

y %

pH

(a) Optimization of Medium pH for TnBglA

Enzyme Activity Protein Conc.

Page 175: A THESIS TITLED - prr.hec.gov.pk

Results

145

Figure 4.35: Effect of medium pH on the expression of recombinant proteins (a) Optimally TnBglA

expression was observed at pH 7.0. (b) TnBglB optimal expression was obtained at neutral pH, both are

defined as 100% activity to calculate the relative activity. Y-error bars show the standard deviation (±SD) of

parallel three experimental reactions. Probability of all the variables is < 5% significance level which entail

that elected variables are significant at 95% confidence interval.

4.8.7. Optimal incubation time after induction

After induction, the incubation time period at which cloned host cell growth, thermophilic

enzymes TnBglA and TnBglB showed maximum expression and activities were optimized.

Recombinant enzyme activity and host culture production was monitored up to 96 hours

after regular intervals. Both recombinant enzymes TnBglA and TnBglB activity increased

up to 72 h post induction beyond which further increment in culture cell growth and

activities were not detected in any of the culture flasks under study, it remained constant.

Therefore, maximum activity was observed after 72 h of induction at 22°C in LB-medium,

TnBglA and TnBglB expression was analyzed on SDS-PAGE (Figure 4.36a and 4.36b).

The maximum expression was found after 72 h induction period and after that slightly

reduced the activities and expression, completely demonstrated in figure 4.37a and 4.37b.

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

0

20

40

60

80

100

120

5 5.5 6 6.5 7 7.5 8

Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

acti

vit

y %

pH

(b) Optimization of Medium pH for TnBglB

Enzyme Activity Protein Conc.

Page 176: A THESIS TITLED - prr.hec.gov.pk

Results

146

Figure 4.36: Analysis of TnBglA and TnBglB expression after induction at different time interval. (a)

Heterologous expression of TnBglA after different time intervals, induced with 0.5 mM IPTG. Lane M,

Protein marker; Lane IC, Crude control supernatant harboring pET-21a (+) after induction with 0.5 mM

IPTG; Lane CL, Crude cell lysate after 24 h induction; Lane S1, Crude supernatant after 24 h induction; Lane

S2, Crude supernatant after 48 h induction; Lane S3, Crude supernatant after 72 h induction; Lane S4, Crude

supernatant after 96 h induction. (b) Expression of extracellular fractions of TnBglB sample withdrawn after

24 h, 48 h and 72 h incubation time, induced with 0.5 mM IPTG. Lane M, Protein marker; Lane 1, Crude

control supernatant harboring pET-21a (+) induced with IPTG; Lane 2, Crude supernatant after 24 h

induction; Lane 3, Crude supernatant after 48 h induction; Lane 4, Crude supernatant after 72 h induction;

Lane 5, Crude supernatant after 96 h induction.

51.5 kDa

81 kDa

25

35

50

(b)

kDa

kDa 225

150

75

50

35

25

100

75

150

(a)

Page 177: A THESIS TITLED - prr.hec.gov.pk

Results

147

Figure 4.37: Effect of incubation time on recombinant enzymes expression (a) After 72 h of induction

maximum TnBglA expression was observed at 22°C with 0.5 mM IPTG induction. (b) Similarly, optimal

expression of TnBglB was found after 72 h of induction at 22°C temperature (0.5 mM IPTG induction), both

are defined as 100% activity to calculate the relative activity. Y-error bars show the standard deviation (±SD)

of parallel three independent experimental reactions. Probability of all the variables is < 5% significance

level which entail that elected variables are significant at 95% confidence level.

0

1

2

3

4

5

0

20

40

60

80

100

120

6 12 18 24 30 36 42 48 54 60 66 72 78 84 90 96

Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

acti

vit

y %

Time (h)

(a) Optimization of Incubation Time for TnBglA Expression

Enzyme Activity Protein Conc.

0.5

1

1.5

2

2.5

3

3.5

4

4.5

5

0

20

40

60

80

100

120

6 12 18 24 30 36 42 48 54 60 66 72 78 84 90 96

Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

acti

vit

y %

Time (h)

(b) Optimization of Induction Time for TnBglB

Enzyme Activity Protein Conc.

Page 178: A THESIS TITLED - prr.hec.gov.pk

Results

148

4.9. Purification of Recombinant TnBglA and TnBglB Proteins

For the purification of soluble extracellular unrefined recombinant TnBglA and TnBglB

proteins, positive bacterial transformants harboring the pET-21a–TnbglA and pET-21a–

TnbglB construct plasmids, were grown individually in fresh sterilized LB-broth medium

(5 L flasks) supplemented with both antibiotic (100 µg mL−1 ampicillin and 50 µg mL-1

chloramphenicol). The recombinant enzymes were grown at optimal cultivation conditions

such as production in neutral pH media, induced culture at 0.6 OD600nm with reduced

concentration of IPTG (0.5 mM) after heat shock treatment at 42°C for 60 minutes followed

by incubated at 22°C for 72 h in a shaking incubator (200 rev min-1). Unrefined cloned

enzymes TnBglA and TnBglB were purified by employing ion exchange (IEX) and

hydrophobic interaction (HIC) chromatography techniques after heat precipitation,

ammonium sulfate fractionation and dialysis. Both recombinant TnBglA and TnBglB

enzymes were efficaciously purified to homogeneity with single bands on 12% SDS-

PAGE. However, activities of both enzymes were monitored at each step of the purification

process, 7.28 fold (40.8% yield) and 7.39 fold purification (39.5% yield) were achieved,

respectively.

All results of purification steps were discussed in the following section.

4.9.1. Heat Treatment

Transformed host cells of E. coli BL21 CodonPlus were harvested from the culture after

72 h induction period, TnBglA and TnBglB expressions and catalytic activities were

detected in all fractions, and the maximum activities were observed with the extracellular

fraction. Therefore, stockpiled the both culture supernatants and concentrated twenty folds

on a lyophilizer or freeze dryer. After that enzyme samples was subjected to water bath at

70°C for 60 minutes, and enriched samples centrifuged (10,000×g) to remove all denatured

host heat labile proteins. After heat treatment, both TnBglA and TnBglB enzymes samples

were run on the SDS-PAGE to analyze the cloned protein purity, most of the mesophilic

host proteins were eliminated which proved it was an effective way to purified

thermostable recombinant protein (Figure 4.38a and 4.38b).

Page 179: A THESIS TITLED - prr.hec.gov.pk

Results

149

4.9.2. Ammonium sulfate precipitation and Dialysis

Followed by heat treatment, the enzyme samples were subjected to precipitation with 55–

60% ammonium sulfate saturation to remove denatured host proteins, collected all

fractional pellets followed by dialysis the resultant suspension to remove (NH4)2SO4 salt

from the precipitated protein samples.

4.9.3. Anion Exchange Chromatography (AEC)

Both recombinant enzymes (TnBglA and TnBglB) were subjected to anion exchange

chromatography, individually. The sample aliquot was loaded to a Resource Q anion

exchange column (Amersham Pharmacia Biotech, Sweden) after dialysis, and active bound

protein fractions were eluted efficiently with a 0-1 M NaCl gradient. According to peak,

collected fractions were analyzed separately, by activity assay and SDS-PAGE after that

active fractions pooled were together. After anion exchange column (AEC)

chromatography, both TnBglA and TnBglB were almost purified but to make desired

protein samples completely purified, the pooled fractions sample was loaded to Resource

ISO pre-packed high performance hydrophobic interaction column (Amersham Pharmacia

Biotech, Sweden).

4.9.4. Hydrophobic Interaction Chromatography (HIC)

For further purification, pooled active fraction sample was loaded on hydrophobic

interaction column (HIC) chromatography individually for both recombinant enzymes. The

bound enzyme fractions were eluted with a 1.5-0 M (NH4)2SO4 decreasing gradient;

various fractions were collected across the major peak. Active enzyme was obtained as

unbound fractions, pooled all active fractions of recombinant enzymes. The final active

fractions of recombinant TnBglA and TnBglB proteins were subjected to SDS-PAGE

analysis, a single prominent band revealed that the target heterologous TnBglA and TnBglB

protein was successfully purified (Figure 4.38a and 4.38b). Recombinant enzymes

activities were monitored at each step of the purification process, 7.28 fold with a recovery

of 40.8% yield and 7.39 fold purification with a recovery of 39.5% yield were achieved,

respectively (Table 4.7a and 4.7b).

Page 180: A THESIS TITLED - prr.hec.gov.pk

Results

150

Figure 4.38 (a): SDS-PAGE analysis of TnBglA from pET-21a–TnbglA in E. coli BL21 CodonPlus (DE3)-

RIPL. Lane M, Novagen Protein marker (Cat # 69079 Lane -3); Lane 1: Crude control supernatant of E. coli

BL harboring pET-21a(+) induced with IPTG (0.5 mM); Lane 2, Crude control supernatant of E. coli

harboring pET-21a(+) without IPTG induction; Lane 3, Crude TnBglA supernatant of E. coli harboring pET-

21a–TnbglA without induction; Lane 4, Crude TnBglA supernatant of E. coli harboring pET-21a(+)–TnbglA

induced with IPTG (0.5 mM); Lane 5, Crude supernatant of TnBglA after heat treatment (at 70°C for 1 h);

Lane 6, purified TnBglA after AEC and HIC (3 µg).

Table 4.7 (a): Purification summary of recombinant β-1,4-glucosidase from T. naphthophila (TnBglA).

Purification steps

Total

protein

(mg)

Total

activity

(kU)

Specific

activity

(kU mg-1)

Yield

(%)

Purification

(fold)

Crude extract 16.4 574 35 100 1.0

Heat treatment and (NH4)2SO4

precipitation 3.16 339.7 107.5 59.2 3.07

Ion-exchange and hydrophobic

interaction chromatography 0.92 234.6 255 40.8 7.28

Each value represent the average of triplicate measurements and varies from the mean by not more than 10%.

kDa

50

35

25

75

100

150

225

51.5 kDa

Page 181: A THESIS TITLED - prr.hec.gov.pk

Results

151

Figure 4.38 (b): SDS-PAGE analysis of TnBglB from pET-21a–TnbglB in E. coli BL21 CodonPlus (DE3)-

RIPL. Lane M, Novagen Protein marker (Cat # 69079-3); Lane 1: Crude control supernatant of E. coli

harboring pET-21a(+) induced with IPTG (0.5 mM); Lane 2, Crude control supernatant of E. coli harboring

pET-21a(+) without induction; Lane 3, Crude TnBglB supernatant of E. coli harboring pET-21a– TnbglB

without induction; Lane 4, Crude TnBglB supernatant of E. coli harboring pET-21a(+)–TnbglB induced with

IPTG (0.5 mM); Lane 5, Crude supernatant of TnBglB after heat treatment (at 70°C for 1 h); Lane 6, purified

TnBglB after AEC and HIC (3 µg).

Table 4.7 (b): Purification summary of recombinant β-1,4-glucosidase from T. naphthophila (TnBglB)

Purification steps

Total

protein

(mg)

Total

activity

(kU)

Specific

activity

(kU mg-1)

Yield

(%)

Purification

(fold)

Crude extract 16.8 295.68 17.6 100 1.0

Heat treatment and (NH4)2SO4

precipitation 3.15 176.4 56 59.6 3.18

Ion-exchange and hydrophobic

interaction chromatography 0.90 117 130 39.5 7.38

Each value represent the average of triplicate measurements and varies from the mean by not more than 10%.

kDa

225 150

100

25

35

50

75

81 kDa

Page 182: A THESIS TITLED - prr.hec.gov.pk

Results

152

4.9.5. Native-PAGE analysis

The purified TnBglA and TnBglB subjected to native-PAGE analysis under non-reducing

conditions, with and without boiling for 10 minutes. Recombinant purified TnBglA

remained as a monomeric form even after boiling for 10 minutes; similarly, TnBglB

remained as a trimeric form even after boiling for 10 minutes which demonstrated that

TnBglB was a homotrimer, with a monomeric molecular weight of 81.1 kDa (Figure 4.39a

and 4.39b).

Figure 4.39: Native PAGE analysis of TnBglA and TnBglB. (a) Lane 1, monomeric recombinant TnBglA (3

µg) without boiling, represented a single prominent band; Lane 2, monomeric TnBglA (3 µg) form displayed

a single band even after 10 minutes boiling at 100°C. (b) Lane 1, homotrimeric recombinant TnBglB (3 µg)

with a single band without boiling; Lane 2, homotrimeric form with a single band of TnBglB (3 µg) even

after 10 minutes boiling.

(a) (b)

Page 183: A THESIS TITLED - prr.hec.gov.pk

Results

153

4.10. Characterization of Purified TnBglA and TnBglB

The complete characterization of recombinant purified TnBglA and TnBglB enzymes from

T. naphthophila were done and determined their molecular weight, optimal pH and

temperature, thermal and pH stability, substrate specificity, kinetics parameters,

thermodynamics study of substrate hydrolysis and enzymes inactivation, shelf life of

enzymes, effect of chemical inhibitors, metallic cation, glucose and xylose on the catalytic

efficiency of cloned thermostable enzymes.

4.10.1. Determination of Molecular weight

The final active fractions of recombinant proteins TnBglA and TnBglB exhibited a single

noticeable band of 51.50 kDa and 81 kDa molecular weight (MW) by 12% SDS-PAGE

analysis (Figure 4.38a and 4.38b). Theoretical molecular weight of cloned enzymes were

50.51 kDa and 81.14 kDa, deduced from amino acid sequence of protein, which revealed

the target that heterologous TnBglA and TnBglB enzymes was successfully purified.

4.10.2. Buffer system and pH profile of TnBglA and TnBglB

Various buffering systems were used to evaluate the optimal buffer for the purified

recombinants TnBglA and TnBglB enzymes, the results were quite similar as experienced

with crude recombinant enzymes and the best buffer system was McIlvaine buffer (for pH

3.0-7.0), HEPES buffer (for pH 8.0) and CAPS buffer (for pH 9.0-11.0), while relatively

reduced hydrolytic efficiency was observed in Tris-Cl buffer, MES buffer, Sorensen’s

phosphate buffer, and MOP buffer. Purified TnBglA and TnBglB enzymes exhibited

maximum activity at pH 7.0 and pH 5.0 McIlvaine buffer, respectively. TnBglA retained

more than 90% of optimal activity over a broad pH range of 6.0-9.0 (Figure 4.40a), whereas

activity was completely repressed at pH 3.0-4.0 and at pH 10.0-11.0. TnBglB exhibited

90% catalytic activity at acidic pH 4.0, retained more than 70% efficiency in McIlvaine

buffer at pH 6.5 and completely repressed in alkaline aqueous condition (Figure 4.40b).

Page 184: A THESIS TITLED - prr.hec.gov.pk

Results

154

Figure 4.40: Effect of pH on the activity of purified TnBglA and TnBglB from T. naphthophila. pH profile

was determined at 70°C for 10 min incubation against pNPG (4 mg mL-1) as substrate, at different pH values

ranging from 3.0-11.0 using McIlvaine buffer (closed square, pH 3.0-7.0), HEPES (closed triangle, pH 8.0)

and CAPS (closed circle, pH 9.0-11.0) buffers. (a) The maximum TnBglA activity observed at pH 7.0, while

(b) TnBglB exhibited maximum activity at pH 5.0, both are defined as 100% activity to calculate the relative

activity. All values shown in results are the average of triplicate experiments. Y-error bars indicate standard

deviation (+SD) among the triplicate experimental reactions, which differ significantly at p≤0.05.

0

20

40

60

80

100

120

3 4 5 6 7 8 9 10 11

Rel

ati

ve

act

ivit

y %

pH

(a) Effect of pH on TnBglA activity

0

20

40

60

80

100

120

3 4 5 6 7 8 9 10 11

Rel

ativ

e ac

tivi

ty %

pH

(b) Effect of pH on TnBglB activity

Page 185: A THESIS TITLED - prr.hec.gov.pk

Results

155

4.10.3. Temperature profile

Optimal temperature of purified enzymes were scrutinized by incubating assay for 10 min

at different temperature (30-100°), pNPG (4 mg mL-1) used as a substrate. TnBglA and

TnBglB displayed peak catalytic activity at 95°C (pH 7.0) and 85°C (pH 5.0), respectively.

TnBglA retained more than 80% of maximum activity over a range of temperature at 80-

100°C, while TnBglB showed more than 75% activity at 70-95°C (Figure 4.41a and 4.41b).

Figure 4.41: Effect of temperature on the activity of purified TnBglA and TnBglB. Temperature profile was

determined, at various temperature range from 30-100°C using pNPG (4 mg mL-1) as substrate, by incubating

TnBglA and TnBglB enzymes for 10 minutes in McIlvaine buffer pH 7.0 and pH 5.0, respectively. (a) TnBglA

displayed optimal activity at 95°C while (b) TnBglB showed optimal catalytic activity at 85°C, both are

defined as 100% activity to calculate the relative activity. Reported data is the average of at least three

independent experiments with the standard deviation (+ SD) presented as error bars, which differ

significantly at p≤0.05.

0

20

40

60

80

100

120

30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105

Rel

ativ

e ac

tiv

ity

%

Temperature °C

(a) Effect of Temperature on TnBglA activity

0

20

40

60

80

100

120

30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105

Rel

ativ

e ac

tivi

ty %

Temperature °C

(b) Effect of Temperature on TnBglB activity

Page 186: A THESIS TITLED - prr.hec.gov.pk

Results

156

4.10.4. Optimal Incubation Time

Time course profile of recombinant TnBglA and TnBglB enzymes for the hydrolysis of

pNPG substrate were monitored, and the optimal incubation time for TnBglA at 95°C

(using buffer pH 7.0) and TnBglB at 85°C (using buffer pH 5.0) was 10 minutes. Both

enzymes slowly increased the catalytic activity and after 10 minute 100% activity was

observed (Figure 4.42).

Figure 4.42: Effect of time course profile of recombinant enzymes. TnBglA and TnBglB displayed optimal

catalytic activity after 10 minutes of incubation at 95°C and 85°C, respectively. Both are defined as 100%

activity to calculate the relative activity. Reported values in results are the average of three independent

experiments, Y-error bars show the standard deviation (±SD) among three reactions, which differ

significantly at p≤0.05.

4.10.5. Thermal stability

Thermal stability of purified TnBglA and TnBglB revealed that both recombinant enzymes

showed remarkable thermotolerant behavior, at pH neutral or near neutral conditions. Both

purified enzymes were incubated in various pH buffers ranging from pH 4.0-7.5 at different

temperature (30-100°C), the enzyme sample was withdrawn at regular interval followed

by activity assay. Purified TnBglA and TnBglB enzymes showed great thermal stability

ranging from 30-85°C temperature at pH 6.5 to 7.5, whereas in acidic conditions (pH 4.0-

5.5) both recombinant enzymes were less stable at 30-85°C as compared to neutral

environment. TnBglA was completely stable after 720 minutes (12 h) incubation at 85°C

in pH 6.5 and 7.5 buffering conditions, while purified TnBglB enzyme showed 100%

0

20

40

60

80

100

120

0 5 10 15

Rel

ativ

e ac

tivi

ty %

Time (minute)

Effect of Incubation Time for Activity Assay

TnBglA TnBglB

Page 187: A THESIS TITLED - prr.hec.gov.pk

Results

157

stability at 85°C after 480 minutes (8 h) incubation in neutral aqueous environment.

TnBglA lost its 50% original activity after 120 incubation at 100°C (Figure 4.43a) whereas,

TnBglB lost its 45% original (optimal) hydrolytic efficiency after 30 minutes incubation at

100°C (Figure 4.43b).

Figure 4.43: Thermal stability of TnBglA and TnBglB at neutral aqueous environment. (a) Thermostability

of TnBglA was determined by pre-incubating purified TnBglA enzyme at temperature ranging from 85-100°C

for different time intervals. (b) Thermostability of TnBglB was determined by pre-incubating purified

TnBglB enzyme at temperature ranging from 80-100°C for different time intervals. Reported values in results

are the average of three independent experiments, Y-error bars show the standard deviation (±SD) among

three reactions, which differ significantly at p≤0.05.

0

20

40

60

80

100

120

0 60 120 180 240 300 360 420 480 540 600 660 720

Rel

ativ

e a

ctiv

ity

%

Time (min)

(a) Thermostability of TnBglA

85°C 90°C 95°C 100°C

0

20

40

60

80

100

120

0 60 120 180 240 300 360 420 480 540 600 660 720

Rel

ativ

e ac

tivi

ty %

Time (min)

(b) Thermostability of TnBglB

85°C 90°C 95°C 100°C

Page 188: A THESIS TITLED - prr.hec.gov.pk

Results

158

4.10.6. pH Stability

Aqueous environment with different pH, greatly affect the enzyme stability, TnBglA and

TnBglB exhibited high pH stability when incubated purified enzymes at 70°C in different

pH buffers ranging from pH 3.0-11.0 for 60 minutes. TnBglA retained more than 90%

residual activity from pH 6.0-9.0 (Figure 4.44a). While, TnBglB retained 100% of its

original activity at pH 6.5-7.5 and more than 50% of its original activity at was observed

pH 4.0-5.5 (Figure 4.44b). Hence, both enzymes were more stable at neutral pH condition

as compared to the acidic and alkaline aqueous conditions.

Figure 4.44: pH stability (a) TnBglA and (b) TnBglB was determined by pre-incubating enzyme at different

pH values (3.0-11.0) at 70°C for 60 minutes, using McIlvaine buffer (closed square, pH 3.0-7.0), HEPES

(closed triangle, pH 8.0) and CAPS (closed circle, pH 9.0-11.0) buffers, activity of enzyme without pre-

incubation is taken as 100%. Y-error bars indicate standard deviation (+SD) among the triplicate

experimental reactions, which differ significantly at p≤0.05.

0

20

40

60

80

100

120

3 3.5 4 4.5 5 5.5 6 6.5 7 7.5 8 8.5 9 9.5 10 10.5 11 11.5

Rel

ativ

e ac

tivi

ty %

pH

(a) pH stability of TnBglA

0

20

40

60

80

100

120

3 3.5 4 4.5 5 5.5 6 6.5 7 7.5 8 8.5 9 9.5 10 10.5 11 11.5

Rel

ativ

e ac

tivi

ty %

pH

(b) pH stability of TnBglB

Page 189: A THESIS TITLED - prr.hec.gov.pk

Results

159

4.10.7. Shelf life of recombinant TnBglA and TnBglB

Storage stability of both enzymes was an important and crucial parameter for commercial

utilization, analyzed in crude media supernatant, after heat precipitation (70°C for 1 h) and

purified samples. TnBglA and TnBglB enzymes had an exceptional storage stability,

retaining 100% initial activity after the storage for two year (till now) at 4°C, in all the

three ways of storage (crude supernatant, heat treated and purified). Heat treated and

purified TnBglA and TnBglB had retained 100% initial activity at room temperature up to

175 days but after 35 days the crude supernatant enzymes showed marginal decline of 5-

7%. Both enzymes showed similar shelf life stability.

4.10.8. Effect of metallic ions and different chaotropic agents

Generally industrial enzymes cannot perform their catalytic work proficiently in the

presence of metallic cations, detergents and chemical inhibitors, because the toxic metallic

cations and chemical reagents interact and alter the enzymes activity in different ways. The

manipulation of metal ions (10 mM) on TnBglA and TnBglB activities were only nominal,

and exhibited optimal hydrolytic efficiency even by the addition of different tri-/divalent

metallic cations. Although Mn2+, Zn2+ , Co2+ and Ca2+ stimulate the activity of TnBglB up

to 115%, 120%, 110% and 125%, respectively. Only Hg2+ and SDS effectively inhibited

the TnBglA and TnBglB activities up to certain limit. However EDTA, Urea, Triton X-100,

Tween-80 and β-mercaptoethanol did not show any conspicuous influence on TnBglA and

TnBglB enzymes activities (Figure 4.45a and 4.45b).

Page 190: A THESIS TITLED - prr.hec.gov.pk

Results

160

Figure 4.45: Effect of metal ions and different inhibitors on purified TnBglA and TnBglB activity. Residual

enzyme activity was expressed as a percentage with pNPG (4 mg mL-1) as substrate, under standard assay

conditions after the addition of metal ions (10 mM), EDTA (10 mM) and chemical reagents (1%) in reaction

mixture (a) TnBglA did not show any significant influence in the presence of inhibitors, while Hg2+ and SDS

detergent cause an obvious inhibitory effect. (b) TnBglB did not reveal any significant influence in the

presence of inhibitors, although Zn2+ and Co2+ cause simulative effect, only Hg2+ and SDS inhibited the

enzyme activity. Y-error bars specify the standard deviation (±SD) of parallel triplicate experimental

reactions. Probability of all the variables is < 5% significance level which entail that elected variables are

significant at 95% confidence interval.

0

20

40

60

80

100

120

Con

tro

l

Ca²⁺

Ni²⁺

Zn²⁺

Mn²⁺

Pb²⁺

Cd²⁺

Co²⁺

Fe²⁺

Mg²⁺

Cu²⁺

Hg²⁺

ED

TA

SD

S

Ure

a

Tw

een

-80

Tri

ton-

X-1

00

β-M

erca

pto

etha

nol

Rel

ati

ve

act

ivit

y %

Metal ions (10 mM), EDTA (10 mM) and chemical reagents (1%)

(a) Effect of Metal ions, chelating agent and chemical reagents on TnBglA

0

20

40

60

80

100

120

140

160

Con

tro

l

Ca²⁺

Ni²⁺

Zn²⁺

Mn²⁺

Pb

²⁺

Cd²⁺

Co²⁺

Fe²⁺

Mg²⁺

Cu²⁺

Hg²⁺

ED

TA

SD

S

Ure

a

Tw

een

-80

Tri

ton-

X-1

00

β-M

erca

pto

etha

nol

Rel

ativ

e ac

tivi

ty %

Metal ions (10 mM), EDTA (10 mM) and chemical reagents (1%)

(b) Effect of Metal ions, chelating agent and chemical reagents on TnBglB

Page 191: A THESIS TITLED - prr.hec.gov.pk

Results

161

4.10.9. Effect of organic solvent

All organic solvents cause a noticeable influence on purified TnBglA and TnBglB enzymes

activity as the concentration increased. Acetonitrile and Ethyl acetate repressed the activity

of purified recombinant TnBglA enzyme up to certain limit. Purified TnBglA activity was

significantly inhibited by addition of 40% (v/v) acetone, ethyl acetate, acetonitrile,

dimethyl sulfoxide (DMSO) and Dimethylformamide (DMF) up to 18%, 38%, 50%, 52%

and 50%, respectively (Figure 4.46a). Whereas, 50% (v/v) acetone, ethyl acetate,

acetonitrile, dimethyl sulfoxide (DMSO) and Dimethylformamide (DMF) influenced the

activity of TnBglB up to 50%, 56%, 54%, 50% and 48% respectively (Figure 4.46b).

4.10.10. Effect of Alcohol

Effect of straight chain aliphatic alcohols (methanol, ethanol, n-butanol and isopropanol)

on TnBglA and TnBglB activity were scrutinized by addition of the test short chain length

alcohols at the final concentration of 2-30% (v/v) in the reaction mixture. These alcohols

have proven an obvious inhibitory additives for the recombinant TnBglA, and drastically

reduced the activity up to 8-10% at the final concentration of 30% (v/v) (Figure 4.47a).

TnBglB showed more resistant behavior as compared to TnBglA, these alcohols did not

exhibit any significant influence on the activity of purified TnBglB at the final

concentration of 20-25% (v/v), whereas at high concentration of 50% (v/v) methanol,

ethanol, n-butanol and isopropanol repressed the TnBglB activity up to 24%, 26%, 22%

and 25%, respectively (Figure 4.47b).

Page 192: A THESIS TITLED - prr.hec.gov.pk

Results

162

Figure 4.46: Effect of organic solvent (acetone, ethyl acetate and acetonitrile) on purified TnBglA and

TnBglB activity. Residual enzyme activity was determined using pNPG (4 mg mL-1) as substrate, under

standard assay conditions and expressed as a percentage (a) TnBglA significant influence in the presence of

50% organic solvents. (b) Organic solvents slightly less inhibited TnBglB activity. Y-error bars specify the

standard deviation (±SD) of parallel triplicate experimental reactions. Probability of all the variables is < 5%

significance level which entail that elected variables are significant at 95% confidence interval.

0

20

40

60

80

100

120

0 5 10 20 30 40 50

Rel

ati

ve

Act

ivit

y %

% Reagent (v/v)

(a) Effect of Organic Solvent on TnBglA activity

Acetone Ethyl acetate Acetonitrile DMSO DMF

0

20

40

60

80

100

120

0 5 10 20 30 40 50

Rel

ativ

e A

ctiv

ity

%

% Reagents (v/v)

(b) Effect of Organic Solvent on TnBglB activity

Acetone Ethyl acetate Acetonitrile DMSO DMF

Page 193: A THESIS TITLED - prr.hec.gov.pk

Results

163

Figure 4.47: Effect of short chain alcohol (ethanol, methanol and isopropanol) on purified TnBglA and

TnBglB activity. Residual enzyme activity was expressed as a percentage with pNPG (4 mg mL-1) as

substrate, under standard assay conditions (a) TnBglA showed obvious constrain in the presence of short

chain alcohol. (b) TnBglB did not displayed any significant influence in the presence of 0-20% alcohols. Y-

error bars specify the standard deviation (±SD) of parallel triplicate experimental reactions. Probability of all

the variables is < 5% significance level which entail that elected variables are significant at 95% confidence

level.

0

20

40

60

80

100

120

0 2 5 10 15 20 25 30

Rel

ati

ve

Act

ivit

y %

% Alcohol (v/v)

(a) Effect of short chain alcohol on purified TnBglA

Ethanol Methanol Isopropanol n-Butanol

0

20

40

60

80

100

120

140

160

0 2 5 10 20 30 40 50

Rel

ativ

e A

ctiv

ity

%

% Alcohol (v/v)

(b) Effect of short chain alcohol on purified TnBglB

Ethanol Methanol Isopropanol n-Butanol

Page 194: A THESIS TITLED - prr.hec.gov.pk

Results

164

4.10.11. Effect of glucose and xylose

Mostly, glucose cause an inhibitory effect on β-glucosidases, to reconnoiter the effect of

glucose and xylose on purified TnBglA and TnBglB activities, various concentration of

glucose and xylose (20-1500 mM) was used to find the inhibition constant (Ki) values of

glucose and xylose for both enzymes. TnBglA catalytic activity was enhanced 160% and

148% by the addition of 100 mM and 200 mM glucose (at final concentration in reaction

mixture), respectively and enzyme remained active even in the presence of 600 mM

glucose. However, TnBglA activity was gradually repressed with the increase

concentration of glucose, and Ki value for glucose was found to be 1200 mM (Figure

4.48a). Similar behavior was observed with the addition of xylose, TnBglA activity was

increased 170% and 154% by adding 100 mM and 200 mM xylose, respectively. TnBglA

catalytic efficiency was gradually repressed with the increase concentration of xylose,

whereas the Ki value was 1300 mM (Figure 4.48b). While, TnBglB enzyme did not show

any stimulatory effect in the presence of glucose and xylose, the relative activity declined

continuously with increased concentration of glucose and its relative activity was repressed

to 12% and 18% in the presence of 600 mM glucose and xylose, respectively. The Ki value

of TnBglB for glucose inhibition was estimated 150 mM, whereas Ki value for xylose was

200 mM (Figure 4.48a and 4.48b).

0

20

40

60

80

100

120

140

160

180

0 200 400 600 800 1000 1200 1400 1600

Rel

ativ

e ac

tivi

ty %

Glucose concentration (mM)

(a) Effect of glucose on TnBglA and TnBglB activity

TnBglA TnBglB

Page 195: A THESIS TITLED - prr.hec.gov.pk

Results

165

Figure 4.48: Effect of glucose and xylose on purified TnBglA and TnBglB activity. (a) Influence of different

glucose concentration (20-1500 mM) on TnBglA and TnBglB activity. (b) Effect of different concentration

of xylose (20-1500 mM) on TnBglA and TnBglB catalytic activity using pNPG (4 mg mL-1) as substrate. Y-

error bars specify the standard deviation (±SD) of parallel triplicate experimental reactions. Probability of all

the variables is < 5% significance level which entail that elected variables are significant at 95% confidence

interval.

4.10.12. Substrate Specificity of TnBglA and TnBglB

Substrate specificity of recombinant TnBglA and TnBglB was examined with various para-

and ortho-nitrophenyl glycoside and heteroglycans substrates. Recombinant highly

thermo-efficient TnBglA displayed great affinity towards pNP-β-D-glucopyranoside

(pNPG) and pNP-β-D-fucopyranoside (pNPF), with highest ever reported specific

activities of 255 kU mg-1 and 253 kU mg-1, respectively. Hydrolytic activity was also

observed with pNP-β-D-xylopyranoside (pNPX, 11 kU mg-1), pNP-β-D-cellobioside

(pNPC, 16 kU mg-1), pNP-β-D-galactopyranoside (pNPGal, 102 kU mg-1) and cellobiose

(177.78 U mg-1). Similarly, TnBglB also showed maximum affinity towards pNPG with

high specific activity of 130 kU mg-1. TnBglB catalytic activity was also found with pNPX

(26 kU mg-1), pNPF (10 kU mg-1), pNPC (8 kU mg-1), pNPGal (7 kU mg-1) and cellobiose

with 150 U mg-1. However, TnBglA and TnBglB were not able to cleave o-nitrophenyl

glycoside substrates, sucrose, carboxymethyl cellulose (CMC), avicel, laminarin and xylan

(Table 4.8).

0

20

40

60

80

100

120

140

160

180

0 200 400 600 800 1000 1200 1400 1600

Rel

ati

ve

act

ivit

y %

Xylose concentration (mM)

(b) Effect of xylose on TnBglA and TnBglB activity

TnBglA TnBglB

Page 196: A THESIS TITLED - prr.hec.gov.pk

Results

166

Table 4.8: Substrate specificity of the recombinant β-glucosidases (TnBglA and TnBglB)

Substrates

TnBglA TnBglB

Specific Activity

(kU mg-1)

Relative

activity

(%)

Specific Activity

(kU mg-1)

Relative

activity

(%)

pNP-β-D-glucopyranoside (pNPG) 255 + 0.12 100 130 + 0.08 100

pNP-α-D-glucopyranoside (pNPαG) 0.005+ 0.14 0.0196 0.004 + 0.03 0.0037

pNP-β-D-fucopyranoside (pNPF) 253 + 0.13 99.2 10 + 0.05 7.7

pNP-α-D-fucopyranoside (pNPF) 0.003+ 0.11 0.012 ND ND

pNP-β-D-galactopyranoside (pNPGal) 102 + 0.03 40 7 + 0.06 5.4

pNP-α-D-galactopyranoside (pNPαGal) ND ND ND ND

pNP-β-D-xylopyranoside (pNPX) 16 + 0.03 6.27 26 + 0.1 10.2

pNP-β-D-cellobioside (pNPC) 11 + 0.19 4.3 8 + 0.04 3.14

pNP-α-D-cellobioside (pNPαC) ND ND ND ND

pNP-β-D-glucuronide (pNPGn) ND ND ND ND

pNP-β-D-mannoside (pNPM) ND ND ND ND

pNP-α-L-arabinofuranoside (pNPαA) ND ND ND ND

p-NP-α-L-fucopyranoside (pNPαLF) ND ND ND ND

oNP-β-D-galactopyranoside (oNPGal) ND ND ND ND

oNP-β-D-fucopyranoside (oNPF) ND ND ND ND

oNP-β-D-glucopyranoside (oNPG) 0.09 + 0.02 0.035 0.6 + 0.03 0.46

Cellobiose 0.177 + 0.001 0.07 0.15 + 0.002 0.12

Sucrose ND ND ND ND

Carboxymethyl cellulose (CMC) ND ND ND ND

Avicel ND ND ND ND

Laminarin ND ND ND ND

Xylan from Birch wood ND ND ND ND

Xylan from Beech wood ND ND ND ND

To analyzed TnBglA and TnBglB catalytic activity, different substrates were tested in McIlvaine buffer

having pH 7.0 at 95°C and pH 5.0 at 85°C, respectively. All data represents the average of three experiments

done in triplicate and the + represents the standard deviation. ND means not detected, specific activity is not

detected by the analytical methods used in this study.

Page 197: A THESIS TITLED - prr.hec.gov.pk

Results

167

4.10.13. Kinetic studies for the hydrolysis of various substrates

Kinetics parameters (Km, Vmax, kcat, and kcat Km-1) of purified TnBglA and TnBglB for the

hydrolysis of various substrates such as pNPG, pNPF, pNPGal, pNPX, pNPC and

cellobiose were analyzed using Lineweaver-Burk plot under standard assay conditions, to

investigate the recombinant enzymes efficiency towards these substrates (Figure 4.49a and

4.49b). Reactions rate were determined by using various concentration of substrates during

activity assay. All kinetic parameters for each substrate that were computed from their

respective Lineweaver-Burk plot and both recombinant TnBglA and TnBglB enzymes

exhibited distinctive affinity towards substrates, results are demonstrated in table 4.9a and

4.9b. Purified TnBglA and TnBglB displayed high Km value 7.76 mM and 7.53 mM against

cellobiose, respectively. Whereas, TnBglA revealed lowest Km value (1.07 mM) with

pNPC, while TnBglB showed lowest Km value (0.45) with pNPG. The highest Vmax 297

mmol mg-1 min-1 of TnBglA was obtained for pNPG with turnover number 1527778 sec-1,

and similarly TnBglB had maximum Vmax value 153 mmol mg-1 min-1 for pNPG with

turnover number 1214285.7 sec-1. Both TnBglA and TnBglB displayed high specificity

constant (kcat Km-1) for pNPG and lowest for cellobiose.

y = 0.5041x + 0.337R² = 0.9793

y = 0.1254x + 0.0849R² = 0.9786

y = 0.5152x + 0.4773R² = 0.8939

y = 0.1558x + 0.0655R² = 0.9821

y = 0.5781x + 0.3381R² = 0.971

y = 1.6479x + 0.0521R² = 0.9832

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

1.8

2

-1 -0.5 0 0.5 1 1.5 2 2.5 3

1/V

1/[S]

(a) Lineweaver–Burk plot for the hydrolysis of various substrates using TnBglA

pNPG pNPGal pNPC pNPX pNPF Cellobiose

Page 198: A THESIS TITLED - prr.hec.gov.pk

Results

168

Figure 4.49: (a) Lineweaver–Burk plot for the hydrolysis of various substrates using TnBglA (b)

Lineweaver–Burk plot for the hydrolysis of various substrates using TnBglB.

Table 4.9 (a): Kinetic parameters of β-glucosidase from T. naphthophila (TnBglA)

Substrates Km (mM) Vmax (mmol mg-1 min-1) kcat (sec-1) kcat Km-1 (mM-1 s-1)

pNPG 1.5 + 0.24 297 + 0.12 1527778 1018518.67

pNPF 1.71 + 0.23 295.7 + 0.13 1521090 889526.31

pNPGal 1.47 + 0.24 117.8 + 0.03 107767.50 73311.2

pNPX 2.37 + 0.21 15.3 + 0.03 78703.70 33208.3

pNPC 1.07 + 0.35 20.95 + 0.19 605967 566324.3

Cellobiose 7.76 + 0.04 0.263 + 0.001 1352.88 174.34

Concentration of TnBglA [e] = 0.00324 µmol.

The Vmax and Km values were determined by plotting the corresponding substrate concentration versus the

initial velocity for each reaction. All values are the average of two experiments done in triplicate.

y = 0.2933x + 0.6547R² = 0.8258

y = 0.2363x + 0.1007R² = 0.9268

y = 0.1052x + 0.0948R² = 0.9045

y = 0.4569x + 0.341R² = 0.9506

y = 0.2054x + 0.0696R² = 0.948

y = 0.3967x + 0.0535R² = 0.9644

0

0.2

0.4

0.6

0.8

1

1.2

1.4

-1 -0.5 0 0.5 1 1.5 2 2.5

1/V

1/[S]

(b) Lineweaver–Burk plot for the hydrolysis of various substrates using TnBglB

pNPG pNPGal pNPC pNPX pNPF Cellobiose

Page 199: A THESIS TITLED - prr.hec.gov.pk

Results

169

Table 4.9 (b): Kinetic parameters of β-glucosidase from T. naphthophila (TnBglB)

Substrates Km (mM) Vmax (mmol mg-1 min-1) kcat (sec-1) kcat Km-1 (mM-1 s-1)

pNPG 0.45 + 0.24 153 + 0.08 1214285.7 2698413

pNPF 2.95 + 0.23 14.37 + 0.05 114047.6 38660

pNPGal 2.35 + 0.24 9.93 + 0.06 78809.5 33536

pNPX 1.34 + 0.21 29.3 + 0.1 232539.7 173537

pNPC 1.11 + 0.35 10.55 + 0.04 83730.2 75433

Cellobiose 7.35 + 0.04 0.185 + 0.002 1469.8 200

Concentration of TnBglB [e] = 0.0021 µmol

The Vmax and Km values were determined by plotting the corresponding substrate concentration versus the

initial velocity for each reaction. All values are the average of two experiments done in triplicate.

4.10.14. Kinetic studies for pNPG hydrolysis at different temperature

Both recombinant TnBglA and TnBglB enzyme were used to find the various Vmax values

at different temperatures (with the increment of 5°C) for pNPG hydrolysis (Table 4.10a

and 4.10b). Maximum Vmax value of TnBglA and TnBglB was observed at 95°C and 85°C,

respectively.

Table 4.10 (a): Kinetic parameters of TnBglA for the hydrolysis of pNPG substrate.

Temp. (K) Km (mM) Vmax (mmol mg-1min-1) kcat (sec-1) kcat Km-1 (mM-1 s-1)

368 (95°C) 1.5 + 0.24 297 + 0.12 1527778 1018518.67

363 (90°C) 1.55 + 0.21 285 + 0.14 1466049 945838.06

358 (85°C) 1.61 + 0.22 254+ 0.11 1306584 811542.86

353 (80°C) 1.02 + 0.24 192+ 0.09 987654 968288.24

Table 4.10 (b): Kinetic parameters of TnBglB for the hydrolysis of pNPG substrate

Temp. (K) Km (mM) Vmax (mmol mg-1min-1) kcat (sec-1) kcat Km-1 (mM-1 s-1)

358 (85°C) 0.45 + 0.24 153 + 0.08 1214285.7 2698413

353 (80°C) 0.24 + 0.21 130 + 0.09 1031746 4298942

348 (75°C) 0.173 + 0.21 108 + 0.04 857143 4954584

343 (70°C) 0.39 + 0.23 102+ 0.06 809524 2075703

Page 200: A THESIS TITLED - prr.hec.gov.pk

Results

170

4.10.15. Thermodynamic of substrate (pNPG) hydrolysis

Thermodynamic study was conducted for the hydrolysis of pNPG substrate, various Vmax

values at different temperatures for pNPG hydrolysis, were applied to construct Arrhenius

plot by taking In Vmax along Y-axis and 1/T along X-axis (Figure 4.50a and 4.50b). The

relationship of temperature-dependent transition state equilibrium of substrate (pNPG)

hydrolysis was explained by Arrhenius approach. Gibb’s free energy (∆G*), Enthalpy

(∆H*) and entropy (∆S*) were determined for the hydrolysis of pNPG by using purified

TnBglA and TnBglB. Activation energy (Ea) for TnBglA and TnBglB was calculated from

the value of slope of Arrhenius plot, which values were 28.74 kJ mol-1 for TnBglA and

27.07 kJ mol-1 for TnBglB. For TnBglA, thermodynamic parameters like ∆H*, ∆G* and

∆S* were calculated by Eyring’s absolute rate equation for 80°C to 95°C, however at

optimal temperature (95°C) of TnBglA as 25.7 kJ mol-1, 47.24 kJ mol-1 and -58.58 J mol-1

K-1, respectively (Table 4.11a). At 85°C optimal temperature of TnBglB, ∆H*, ∆G* and

∆S* values were 24.09 kJ mol-1, 46.55 kJ mol-1 and -62.74 J mol-1 K-1, respectively (Table

4.11b).

y = -3456.8x + 22.033R² = 0.9757

12

12.1

12.2

12.3

12.4

12.5

12.6

12.7

0.0027 0.00272 0.00274 0.00276 0.00278 0.0028 0.00282 0.00284

ln V

max

1/Temperature (1/K)

(a) Arrhenius Plot for pNPG hydrolysis using TnBglA

Page 201: A THESIS TITLED - prr.hec.gov.pk

Results

171

Figure 4.50: Arrhenius plot of recombinant enzymes from T. naphthophila. (a) Arrhenius plot to calculate

activation energy Ea of recombinant TnBglA for the hydrolysis of pNPG (substrate). (b) Arrhenius plot to

calculate activation energy Ea of recombinant TnBglB for the hydrolysis of pNPG (substrate). Ea= –slope ×

R, where R (gas constant) = 8.314 JK-1 mol-1. Error bars show standard deviation among three observations.

Table 4.11 (a): Thermodynamic parameters of TnBglA for pNP-G hydrolysis. Ea = 28.74 kJ mol-1

Temperature

(K)

Vmax

(mmol mg-1 min-1)

kcat

(sec-1)

ΔH*

(kJ mol-1)

ΔG*

(kJ mol-1)

ΔS*

(J mol-1K-1)

368 (95°C) 297 1527778 25.68 47.24 -58.58

363 (90°C) 285 1466049 25.72 46.66 -57.68

358 (85°C) 254 1306584 25.76 46.31 -57.41

353 (80°C) 192 987654 25.80 46.47 -58.55

Concentration of the enzyme [e] = 0.00324 µmol

y = -3256.1x + 20.998R² = 0.9401

11

11.2

11.4

11.6

11.8

12

12.2

0.00278 0.0028 0.00282 0.00284 0.00286 0.00288 0.0029 0.00292 0.00294

ln V

ma

x

1/Temperature (1/K)

(b) Arrhenius Plot for pNPG hydrolysis using TnBglB

Page 202: A THESIS TITLED - prr.hec.gov.pk

Results

172

Table 4.11 (b): Thermodynamic parameters of TnBglB for pNP-G hydrolysis. Ea = 27.07 kJ mol-1

Temperature

(K)

Vmax

(mmol mg-1 min-1)

kcat

(sec-1)

ΔH*

(kJ mol-1)

ΔG*

(kJ mol-1)

ΔS*

(J mol-1K-1)

358 (85°C) 153 1214285.7 24.09 46.55 -62.74

353 (80°C) 130 1031746 24.13 46.31 -62.83

348 (75°C) 108 857143 24.17 46.18 -63.25

343 (70°C) 102 809523.8 24.22 45.65 -62.48

Concentration of the enzyme [e] = 0.0021 µmol

4.10.16. Thermodynamics stability of TnBglA and TnBglB

Thermodynamic parameters for irreversible thermal inactivation of purified recombinant

TnBglA and TnBglB enzyme were calculated. To evaluate the thermal denaturation, the

purified enzyme TnBglA was incubated at temperatures ranging from 97-99°C (in an

increment of 1°C), nearer to the optimal temperature of TnBglA activity and similarly

TnBglB purified enzyme was incubated at temperatures ranging from 94-96°C (in an

increment of 1°C). Pseudo-first-order plots were applied to assess the thermal inactivation

of TnBglA and TnBglB (Figure 4.51a and 4.51b).

Recombinant enzyme TnBglA was found highly stable and showed half-life (t1/2) of 5.2

minutes at 97°C and TnBglB enzyme showed high stability with a half-life (t1/2) of 4.44

minutes at 94°C. Thermodynamic parameters for denaturation of purified TnBglA enzyme

like enthalpy of denaturation (ΔH*D), free energy of denaturation (ΔG*D) and entropy of

denaturation (ΔS*D) at 97°C were found to be 662.04 kJ mol-1, 110.1 kJ mol-1 and 1.491 kJ

mol-1K-1, respectively (Table 4.12a). All these parameters were determined after

calculating the activation energy EaD for irreversible thermal inactivation of TnBglA by

Arrhenius plot (Figure 4.52a). While, thermodynamic parameters for denaturation of

TnBglB enzyme such as enthalpy of denaturation (ΔH*D), free energy of denaturation

(ΔG*D) and entropy of denaturation (ΔS*D) at 94°C were found to be 283.78 kJ mol-1,

108.69 kJ mol-1 and 0.477 kJ mol-1K-1, respectively (Table 4.12b). These parameters were

determined after calculating the activation energy EaD for irreversible thermal inactivation

of TnBglB by Arrhenius plot (Figure 4.52b).

Page 203: A THESIS TITLED - prr.hec.gov.pk

Results

173

Figure 4.51: Pseudo-first-order plots for irreversible thermal denaturation of recombinant thermostable

enzymes. (a) Pseudo-first-order plots for irreversible thermal denaturation of TnBglA. The purified TnBglA

enzyme was pre-incubated at 94°C, 97°C, 98°C and 99°C in the absence of pNPG substrate, after appropriate

time interval TnBglA activity was determined in McIlvaine buffer (pH 7.0) at 95°C, before this incubating

enzyme on ice for 30 minutes. (b) Pseudo-first-order plots for irreversible thermal denaturation of TnBglB.

The purified TnBglB enzyme was pre-incubated at 92°C, 94°C, 95°C and 96°C in the absence of pNPG

substrate, after appropriate time interval TnBglB activity was determined in McIlvaine buffer (pH 5.0) at

85°C, before this incubating enzyme on ice for 30 minutes. Error bars show standard deviation among three

observations.

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

5

0 5 10 15 20 25 30

In R

esid

ua

l a

ctiv

ity

%

Time (min)

(a) Pseudo-first-order plots of TnBglA

94°C 97°C 98°C 99°C

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

5

0 5 10 15 20 25 30

In R

esid

ual

act

ivit

y %

Time (min)

(b) Pseudo-first-order plots of TnBglB

92°C 94°C 95°C 96°C

Page 204: A THESIS TITLED - prr.hec.gov.pk

Results

174

Figure 4.52: Arrhenius plot for irreversible thermal inactivation of purified recombinant enzymes. (a)

Arrhenius plot to calculate activation energy EaD for irreversible thermal inactivation of TnBglA. (b)

Arrhenius plot to calculate activation energy EaD for irreversible thermal inactivation of TnBglB. Error bars

show standard deviation among three observations.

y = -80000x + 214.08R² = 0.9552

-2.5

-2

-1.5

-1

-0.5

00.002676 0.002681 0.002686 0.002691 0.002696 0.002701

lnK

d

1/T

(a) Arrhenius plot for irreversible thermal inactivation of TnBglA

y = -34500x + 92.317R² = 0.9983

-2

-1.9

-1.8

-1.7

-1.6

-1.5

-1.4

-1.3

-1.2

-1.1

-10.002705 0.00271 0.002715 0.00272 0.002725 0.00273 0.002735

lnK

d

1/T

(b) Arrhenius plot for irreversible thermal inactivation of TnBglB

Page 205: A THESIS TITLED - prr.hec.gov.pk

Results

175

Table 4.12 (a): Thermodynamic parameters for denaturation of recombinant β-1,4- glucosidase

(TnBglA) enzyme. EaD = 665.12 kJ mol-1

Temperature

(K)

kd

(min-1)

Half-life

(min)

ΔH*D

(kJ mol-1)

ΔG*D

(kJ mol-1)

ΔS*D

(kJ mol-1K-1)

370 (97°C) 0.133 5.21 662.04 110.10 1.491

371 (98°C) 0.399 1.74 662.03 107.02 1.496

372 (99°C) 0.656 1.06 662.02 105.78 1.495

Table 4.12 (b): Thermodynamic parameters for denaturation of recombinant β-1,4- glucosidase

(TnBglB) enzyme. EaD = 286.83 kJ mol-1

Temperature

(K)

kd

(min-1)

Half-life

(min)

ΔH*D

(kJ mol-1)

ΔG*D

(kJ mol-1)

ΔS*D

(kJ mol-1K-1)

367 (94°C) 0.156 4.44 283.78 108.69 0.477

368 (95°C) 0.215 3.22 283.83 108.02 0.478

369 (96°C) 0.319 2.24 283.76 107.20 0.479

4.11. Structure Analysis of TnBglA and TnBglB

Secondary and three-dimensional (3-D) structures of TnBglA and TnBglB were analyzed

using various online software. In the following section secondary and 3-D structures were

discussed separately for both proteins.

4.11.1.1. Secondary structure of TnBglA

All details related to the secondary structure of TnBglA was retrieved after submitting the

structure in PDB-format to the PDBsum server, the α-helices, β-sheets and random coils

were clearly evident in the secondary structure of TnBglA (Figure 4.53a). According to

the secondary structure of TnBglA, a chain had 446 amino acid residues, in which 21

helices, 2 sheets, 5 gamma turns, 27 beta turns, 11 strands, 2 beta hairpins, 6 beta bulges,

34 helix-helix interactions and 6 beta-alpha-beta units were present. Majority amino acid

residues were involved in the formation of β-turns and α-helices. The amino acid residues

Page 206: A THESIS TITLED - prr.hec.gov.pk

Results

176

in helix were buried (hydrophobic) therefore, not exposed to the surrounding environment

(aqueous).

Figure 4.53 (a): Secondary structure of TnBglA. Predominance of strands and helices along with 2 beta-

hairpins.

4.11.1.2. Secondary structure of TnBglB

The detail secondary structure of TnBglB was retrieved after submitting the structure in

PDB-format to the PDBsum server, the α-helices, β-sheets and random coils were clearly

evident in the secondary structure of TnBglB, as shown in figure 4.53b. According to the

secondary structure of TnBglB, 721 amino acid residues were arranged in 29 helices, 6

sheets, 10 gamma turns, 58 beta turns, 26 strands, 6 beta hairpins, 7 beta bulges, 35 helix-

helix interactions and 5 beta-alpha-beta units. The amino acid residues in helix were buried

(hydrophobic) therefore, not exposed to the surrounding environment (aqueous).

Page 207: A THESIS TITLED - prr.hec.gov.pk

Results

177

Figure 4.53 (b): Secondary structure of TnBglB. Predominance of strands and helices along with 6 beta-

hairpins.

4.11.2.1. Procheck Analysis for TnBglA

The statistics study of Ramachandran plot of TnBglA contained 446 amino acid residues

displayed in the figure 4.54a, which showed the presence of 92.5% amino acid residues in

most favorable regions, 6.4% residues present in additional allowed regions with only one

amino acid in disallowed region (Table 4.13a). All results related to the TnBglA structure

Page 208: A THESIS TITLED - prr.hec.gov.pk

Results

178

is based on 118 structures analysis of resolution of 2.0 Aͦ (Angstroms) approximately and

R-factor no greater than 20%. A model having good quality, would be estimated to have

more than 90% amino acid residues in the most favoured regions.

Figure 4.54 (a): Ramachandran Plot for TnBglA on Procheck using PDBsum

Table 4.13 (a): Statistics of Ramachandran Plot. Results computed after Procheck analysis of

TnBglA protein structure using PDBsum.

Residues No. of

Residues

Frequency of amino acid residues in

Ramachandran plot (%)

In most favoured regions 359 92.5

In additional allowed regions 25 6.4

In generously allowed regions 3 0.8

In disallowed regions 1 0.3

No. of non-glycine and non-proline 388 100

No. of End amino acid 02 -

No. of glycine 35 -

No. of proline 21 -

Total No. of amino acid 446 -

Page 209: A THESIS TITLED - prr.hec.gov.pk

Results

179

4.11.2.2. Procheck Analysis for TnBglB

Ramachandran plot statistics study of TnBglB that consisted of 721 amino acid residues

showed the presence of 92.3% amino acid residues in most favorable regions, in additional

allowed regions 6.6% residues and in disallowed region 0.3% amino acids residues present

(Table 4.13b). Triangles as shown in figure 4.54b displayed the presence of 60 glycine

residues, and total 37 proline residues were present in the structure. All results related to

the TnBglB structure is based on 118 structures analysis of resolution of 2.0 Aͦ (Angstroms)

approximately and R-factor no greater than 20%. A model having good quality, would be

estimated to have more than 90% amino acid residues in the most favoured regions.

Figure 4.54 (b): Ramachandran Plot for TnBglB on Procheck using PDBsum

Page 210: A THESIS TITLED - prr.hec.gov.pk

Results

180

Table 4.13 (b): Statistics of Ramachandran Plot. Results computed after Procheck analysis of

TnBglB protein structure using PDBsum.

Residues No. of

Residues

Frequency of amino acid residues in

Ramachandran plot (%)

In most favoured regions 575 92.3

In additional allowed regions 41 6.6

In generously allowed regions 5 0.8

In disallowed regions 2 0.3

No. of non-glycine and non-proline 623 100

No. of End amino acid 01 -

No. of glycine 60 -

No. of proline 37 -

Total No. of amino acid 721 -

4.11.3.1. Three Dimensional Structure of TnBglA protein

ExPASy-proteomics (GENO-3-D) explained that TnBglA has (α/β)8 triose phosphate

isomerase (TIM) barrel scaffold, which is one of the key feature of GH family 1 enzymes

especially β-glucosidases, are connected through a chain and gives stability to the protein

structure. The 3-D structure of β-glucosidase (TnBglA) represents an interesting pattern of

conserved structural elements in the way that the outer part of the structural fold contains

alpha helices while inner part contains almost all parallel beta sheets forming the catalytic

cleft. In 3-D structure, glutamic acids (Glu166, Glu351 and Glu405) residues were present in

a pocket shaped structure with hydrophobic amino acid residues at the entrance point

leading to the active site pocket (Figure 4.55a). According to the 3-D structure, TnBglA is

a single domain protein, and has 42% helix (H), 10% β-sheet (E) and 47% loop (C). The

quality of 3-D structure was determined by using RaptorX server, and computed the p-

value, uGDT, SeqID and score after submitting amino acid sequence of TnBglA. Model

structure got 519 score, relative quality of a structure was estimated by p-value, TnBglA

computed small 2.83e-18 value, which showed that structure had higher quality. The value

of uSeqID was higher 430(96) which explained that predicted structure folds were correct.

Page 211: A THESIS TITLED - prr.hec.gov.pk

Results

181

uGDT (unnormalized Global Distance Test) measured the absolute model quality, TnBglA

model retrieved a high 492(110) value, showed that model was fairly good. TnBglA model

displayed solvent accessibility (ACC) values of 46% buried (B), 28% medium (M), and

24% exposed (E).

Figure 4.55 (a): Three-dimensional (3-D) model structure of TnBglA with glutamic acid (Glu) residues

(Glu351 and Glu405) as catalytic nucleophile as well as catalytic proton donor (Glu166). The arrows in the center

shows the catalytic cavity for substrate binding covered by parallel β sheets. The structure was predicted

using an automatic protein modeling sever, GENO-3-D, and results analyzed by PyMOL (molecular system

for visualization and graphics).

4.11.3.1.1. Catalytic Triad of TnBglA

Catalytic triad of TnBglA comprise on three well-conserved catalytic residues which are

Glu166, Glu351 and Glu405, the distance between proton donor catalytic residue Glu166 and

nucleophile active catalytic residue Glu351 is 4.7 Aͦ and the distance between proton donor

Glu166 and catalytic nucleophile Glu405 is 10.1 Aͦ, all the values calculated by using PyMOL

molecular graphics system. Although the distance between Glu351 and Glu405 is 8.6 Aͦ as

shown in figure 4.55b.

Page 212: A THESIS TITLED - prr.hec.gov.pk

Results

182

Figure 4.55 (b): Catalytic triad of TnBglA, distance between Glu166 act as proton donor and nucleophile

active residues Glu351 and Glu405

4.11.3.2. Three Dimensional Structure of TnBglB protein

ExPASy-proteomics (GENO-3-D) explained that TnBglB has three-domain structure,

composed of 721 amino acid residues that are arranged in three domains. These domains

are connected by two linkers (307-320 and 537-599 residues), domain-I (1-306 residues)

has an (α/β)8 barrel structure similar to a TIM barrel, domain-II (321-536 residues) has

five-stranded α/β sandwich (α/β)5, both domains are important because have a direct and

crucial role for the active-site organization. Domain-III (600-721 residues) has a

fibronectin type III (FnIII) fold whose functions is still not clear (unknown) (Figure 4.55c).

All domains of 3-D predicted structure of TnBglB (β-glucosidase) are well organized in

which active site present at the interface of domain-I and II forming the catalytic pocket.

Domain-I has an essential catalytic residue aspartic acid (Asp242) which act as nucleophile

while domain-II contains a (α/β)5 sandwich fold and has an acid/base residue glutamic

acids (Glu458) (Figure 4.55c). The 3-D structure of TnBglB composed of 25% helix (H),

Page 213: A THESIS TITLED - prr.hec.gov.pk

Results

183

19% β-sheet (E) and 55% loop (C). The quality of 3-D structure was determined by using

RaptorX server, and computed parameters such as p-value, uGDT, SeqID and score after

submitting amino acid sequence of TnBglB. Model structure got 729 score, relative quality

of a structure was estimated by p-value, TnBglB computed small 7.40e-22 value, which

showed that structure had high quality. The value of uSeqID was higher 634(88), which

elucidated that predicted structure folds were correct. uGDT (unnormalized Global

Distance Test) estimated the absolute model quality, TnBglB model retrieved a high

629(87) value, showed that model was fairly good. TnBglB model displayed solvent

accessibility (ACC) values of 41% buried (B), 28% medium (M), and 30% exposed (E).

Figure 4.55 (c): 3-D model structure of TnBglB colored by domain, along with catalytic residues. Domain-

I, red color (1-306 residues); Domain-II, cyan color (321-536 residues); Domain-III, orange color (600-721).

The linker (307-320 residues) between domain-I and domain-II is shown in green; the linker (537-599

residues) between domain-II and domain-III is represented in yellow. Catalytic active site present at the

interface of domain-I and domain-II, in a pocket like structure in which aspartic acid residues (Asp242) act as

nucleophile and glutamic acid residue (Glu458) act as catalytic proton donor. The structure was predicted

using an automatic protein modeling sever, GENO-3-D, and conserved domains or coding sequence within

a protein studied by using NCBI, results analyzed by PyMOL (molecular system graphic system).

Page 214: A THESIS TITLED - prr.hec.gov.pk

Results

184

4.11.3.2.1. Catalytic residues of TnBglB

TnBglB consist of well-conserved two catalytic residues Asp242 and Glu458, distance

between the proton donor catalytic residue Glu458 and nucleophile active catalytic residue

Asp242 is 5.8 Aͦ (Figure 4.55d). The distance between catalytic amino acid residues

measured by PyMOL molecular system for visualization and graphics.

Figure 4.55 (d): Catalytic residues of TnBglB, distance between Glu458 act as proton donor and nucleophile

active residues Asp242.

4.11.4.1. Docking Studies of TnBglA

The docking studies of enzyme-substrate interaction by PatchDock generated result give

more than 100 possible solutions of docking. All these solutions were evaluated for their

accuracy by Procheck and Ramachandran plots on PDBSum which revealed their stability

(Laskowski, 2009). Additionally, the analysis of enzyme-substrate complexes were also

performed by studying the ligplot (prediction tool for H-bonds) which showed the possible

non-covalent interaction detail of enzyme with the substrates. One of the best complexes

amongst the first 100, for each substrate-TnBglA docking results, having at least more than

80% residues in favored and allowed regions of Ramachandran plots and also having two

conserved glutamic acid (Glu) residues, one in N-terminal site and the other in middle or

C-terminal site was selected for each of the enzyme-substrate complex. The best docking

Page 215: A THESIS TITLED - prr.hec.gov.pk

Results

185

complex of TnBglA with cellobiose substrate was used to analyze the enzyme substrate

interaction (Figure 4.56a). In this reaction complex Tyr295, Asn165, Glu166, Glu351 and Glu405

active-site residues are involved in making hydrogen bonds with ligand, suggested these

amino acid residues may play a crucial role in the substrate hydrolysis process. The

surrounding amino acids Trp324, Phe414, Trp398, Trp122, Try406, His180, Glu408 and Met322

provide a hydrophobic environment to improve reactivity in the interaction. However,

pNPG substrate molecules in the complex system were coordinately linked with the Met322,

Tyr295, Glu351 and Glu405 active-site residues (Figure 4.56b). This interacting system

surrounded by Glu166, Trp398, Trp324, Trp122, Trp406, His180 and Glu408 involved in

hydrophobicity to enhance reactivity and thermophily. All finest complexes for each

substrate revealed that at least two glutamic acidic (Glu) amino acids present in the active

cleft or catalytic site (Figure 4.56c). One important fact that is evident from all these

docking models is that in each docking models there were at least two or at highest three

glutamic acid residues (Glu166, Glu351 and Glu405) were conserved; and these residues were

observed in close contact to the substrate to perform the hydrolytic function.

(a)

Page 216: A THESIS TITLED - prr.hec.gov.pk

Results

186

(b)

Figure 4.56: Ligplot output for H-bond interactions of the amino acid residues of TnBglA (a) involved in

binding with cellobiose (b) with the p-phenyl sugars as substrates.

Page 217: A THESIS TITLED - prr.hec.gov.pk

Results

187

Figure 4.56 (c): Surface view of 3-D structure of TnBglA, the circle (in red colour) represents the catalytic

cleft for substrate binding.

4.11.4.2. Docking Studies of TnBglB

The docking studies of TnBglB enzyme-substrate interaction by PatchDock generated

result give more than 100 possible solutions of docking. All these solutions were evaluated

for their accuracy by Procheck and Ramachandran plots on PDBSum which revealed their

stability (Laskowski, 2009). Additionally, the analysis of enzyme-substrate complexes

were also performed by studying the ligplot (prediction tool for H-bonds) which showed

the possible non-covalent interaction detail of enzyme with the substrates. One of the best

complexes amongst the first 100, for each substrate-TnBglB docking results, having at least

more than 80% residues in favored regions of Ramachandran plots, and having one

conserved nucleophile aspartic acid (Asp242) residue in domain-I and one conserved

catalytic proton donor glutamic acid (Glu458) residue in domain-II, was selected for each

of the enzyme-substrate complex. Both substrates locked in a tight network of hydrogen

bonds in which active-site residues Asp58, Arg64, Glu458 and Asp242 involved. The side

chain of Ser370 and Try243 may form weak hydrogen bond with substrates (Figure 4.57a and

Page 218: A THESIS TITLED - prr.hec.gov.pk

Results

188

4.57b). All best complexes for each substrate demonstrated that two catalytic residues

present in the active cleft or catalytic pocket (Figure 4.57c). Moreover, evident from all

these docking models, it is predicted that in each docking models there were at least two

catalytic residues Asp242 and Glu458 were conserved; and these residues were observed in

close contact to the substrate to perform the hydrolytic function.

(a)

Page 219: A THESIS TITLED - prr.hec.gov.pk

Results

189

(b)

Figure 4.57: Ligplot output for H-bond interactions of the amino acid residues of TnBglB (a) involved in

binding with cellobiose (b) with the p-phenyl sugars as substrates.

Figure 4.57 (c): Surface view of 3-D structure of TnBglB, the circle (in red colour) shows the catalytic cleft

for substrate binding.

Page 220: A THESIS TITLED - prr.hec.gov.pk

Results

190

4.12. Enhanced Production of Recombinant TnBglA and TnBglB

After purification and characterization, the recombinant TnBglA and TnBglB enzymes

production was further enhanced in a mesophilic E. coli BL21 CodonPlus DE3-(RIPL)

host through various cultivation and induction strategies. To enhance the production of

engineered host cells and over-expression of recombinant enzymes, different inducing

media were used which induced individually with various concentration of IPTG and

lactose. Culture conditions and other parameters were also optimized to achieve maximum

yield of heterologous proteins.

4.12.1. Production and Expression of recombinant enzymes

Ten different inducing media including LB, LB+, ZB, 4×ZB, ZBM, ZYB, 3×ZYB,

ZYBM9, 3×ZYBM9, and M9 with some modifications in their composition were used,

which induced individually either with IPTG (0.1-1.0 mM) or lactose (10-300 mM), and

studied the effect of each medium on both TnBglA and TnBglB expression and activity.

Therefore, 1% of all respective seed cultures of both cloned host strains harboring pET-

21a–TnbglA and pET-21a–TnbglB plasmids were used to inoculate their respective

sterilized media containing both antibiotics. Cultivation conditions and parameters

including pre-induction optical density, agitation, concentration of both inducer,

temperature and time of induction, were again optimized in all modified and complex

inducing media to achieve the maximum yield of heterologous proteins. All experimental

results (from various media) are the means of triplicate experiments. Enzymes percentage

(%) were detected by densitometric scanning of Coomassie stained gel (Syngene, Gene

Tools, UK).

4.12.2. Screening of optimal inducing media for TnBglA and TnBglB

The host cells harboring pET-21a–TnbglA and pET-21a–TnbglB plasmid, were grown in

ten different media (LB, LB+, ZB, ZBM, ZYB, 3×ZYB, 4×ZB, M9, ZYBM9, and

3×ZYBM9) supplemented with ampicillin (100 µg mL-1) and chloramphenicol (50 µg mL-

1) induced individually with IPTG and lactose to screen the best medium for both TnBglA

and TnBglB proteins production and expression in terms of activity. Maximum

Page 221: A THESIS TITLED - prr.hec.gov.pk

Results

191

extracellular TnBglA production and activity was found in 4×ZB modified medium

induced with IPTG (0.5 mM) after 72 h induction at 22°C, seemed to be due to greater

recombinant cell density and dry cell weight. However, lowest culture density and activity

was observed in M9 medium (Figure 4.58a). Similarly, optimal production and TnBglB

extracellular enzyme activity found in 4×ZB modified medium induced with 0.5 mM IPTG

(Figure 4.58b), seemed to be due to greater recombinant cell density and dry cell weight.

Figure 4.58: Effect of media on recombinant enzymes activity induced with 0.5 mM IPTG (a): Effect of

inducing media on recombinant TnBglA activity induced with 0.5 mM IPTG after 72 h incubation at 22°C,

optimal activity was observed in 4×ZB medium and least in M9 medium. (b): Effect of inducing media on

recombinant TnBglB activity after induction with 0.5 mM IPTG at 22°C for 72 h, optimal activity was

observed in 4×ZB medium and least in M9 medium. Y-error bars specify the standard deviation (±SD) of

parallel triplicate experimental reactions. Probability of all the variables is < 5% significant level which entail

that elected variables are significant at 95% confidence interval.

0

20

40

60

80

100

120

LB

LB

+

ZB

4Z

B

ZY

B

3Z

YB

ZY

BM

9

3Z

YB

M9

ZB

M

M9

Rel

ativ

e a

ctiv

ity

%

Media

(a) Optimization of Media Induced with 0.5 mM IPTG for TnBglA

24 h 48 h 72 h

0

20

40

60

80

100

120

LB

LB

+

ZB

4ZB

ZY

B

3Z

YB

ZY

BM

9

3ZY

BM

9

ZB

M

M9

Rel

ativ

e ac

tivi

ty %

Media

(b) Optimization of Media Induced with 0.5 mM IPTG for TnBglB

24 h 48 h 72 h

Page 222: A THESIS TITLED - prr.hec.gov.pk

Results

192

Induced with lactose (150 mM), peak TnBglA activity and cultural density were also

observed in 4×ZB after 72 h as compared with other media (Figure 4.59a). Similarly

maximum TnBglB activity was obtained with 150 mM lactose in 4×ZB medium after 72 h

incubation at optimal conditions as compared with other inducing media (Figure 4.59b).

Figure 4.59: Effect of media on recombinant enzymes activity induced with 150 mM lactose (a): Effect of

inducing media on recombinant TnBglA activity induced with 150 mM lactose after 72 h incubation at 22°C,

optimal activity was observed in 4×ZB medium and least in M9 medium. (b): Effect of inducing media on

TnBglB activity after induction with 150 mM lactose at 22°C for 72 h, optimal activity was observed in 4×ZB

medium and least in M9 medium. Y-error bars specify the standard deviation (±SD) of parallel triplicate

experimental reactions. Probability of all the variables is < 5% significant level which entail that elected

variables are significant at 95% confidence level.

0

20

40

60

80

100

120L

B

LB

+

ZB

4ZB

ZY

B

3Z

YB

ZY

BM

9

3ZY

BM

9

ZB

M

M9

Rel

ati

ve

act

ivit

y %

Media

(a) Optimization of Media Induced with 150 mM Lactose for TnBglA

24 h 48 h 72 h

0

20

40

60

80

100

120

LB

LB

+

ZB

4Z

B

ZY

B

3ZY

B

ZY

BM

9

3ZY

BM

9

ZB

M

M9

Rel

ativ

e ac

tivi

ty %

Media

(b) Optimization of Media Induced with 150 mM Lactose for TnBglB

24 h 48 h 72 h

Page 223: A THESIS TITLED - prr.hec.gov.pk

Results

193

4.12.2.1. Expression of TnBglA in various Media induced with IPTG

The effect of various cultivation media induced with 0.5 mM IPTG, on the expression of

TnBglA was examined by comparing the heterologous protein profiles of recombinant

E.coli BL21 CodonPlus (DE3) cultivated in LB, ZB, ZBM, ZYB, 3×ZYB, 4×ZB, M9,

ZYBM9, and 3×ZYBM9 inducing media (Figure 4.60a and 4.60b).

Figure 4.60: SDS-PAGE analysis of recombinant TnBglA in E. coli BL 21 grown in various 0.5 mM IPTG

inducing media (a) Lane M, Protein marker; Lane C, Crude control supernatant harboring pET-21a (+)

without induction; Lane 1, Crude supernatant proteins of cells cultivated in LB; Lane 2, Crude heat treated

LB; Lane 3, supernatant cultivated in ZB; Lane 4, heat treated supernatant of ZB; Lane 5, supernatant protein

of ZBM; Lane 6, heat treated supernatant of ZBM; Lane 7, Supernatant of 4×ZB; Lane 8, heat treated

supernatant of 4×ZB. (b) Lane C, Crude control supernatant; Lane 1, supernatant protein of ZYB; Lane 2,

heat treated supernatant of ZYB; Lane 3, supernatant of 3×ZYB; Lane 4, heat treated supernatant of 3×ZYB;

Lane 5, supernatant of ZYBM9; Lane 6, heat treated supernatant of ZYBM9; Lane 7, supernatant of

3×ZYBM9; Lane 8, heat treated supernatant of 3×ZYBM9.

kDa

100

TnBglA 51.5 kDa

75

50

35

25

(a)

TnBglA 51.5 kDa

(b)

Page 224: A THESIS TITLED - prr.hec.gov.pk

Results

194

4.12.2.2. Expression of TnBglB in various Media induced with IPTG

Various cultivation inducing media LB, ZB, ZBM, ZYB, 3×ZYB, 4×ZB, M9, ZYBM9,

and 3×ZYBM9 were used to analyzed the effect of growth media induced with 0.5 mM

IPTG on the expression of TnBglB by comparing the heterologous protein profiles of

recombinant E.coli BL21 CodonPlus (DE3) cultivated in all these inducing media (Figure

4.61a and 4.61b).

Figure 4.61: SDS-PAGE analysis of TnBglB in E. coli grown in various IPTG inducing media (a) Lane M,

Protein marker; Lane 1, Control supernatant harboring pET-21a (+) without induction; Lane 2, Crude

supernatant cells proteins cultivated in ZYB; Lane 3, Crude heat treated ZYB; Lane 4, supernatant cultivated

in ZB; Lane 5, heat treated supernatant of ZB; Lane 6, supernatant protein of ZBM; Lane 7, heat treated

supernatant of ZBM; Lane 8, Supernatant of 3×ZYBM9; Lane 8, heat treated supernatant of 3×ZYBM9. (b)

Lane M, Protein marker; Lane 1, Crude control supernatant; Lane 2, supernatant of 4×ZB; Lane 3, heat

treated supernatant of 4×ZB; Lane 4, supernatant of LB; Lane 5, heat treated supernatant of LB; Lane 6,

supernatant of 3×ZYB; Lane 7, heat treated supernatant of 3×ZYB; Lane 8, supernatant of ZYBM9; Lane 9,

heat treated supernatant of ZYBM9.

TnBglB

81 kDa

(a)

75

100 kDa

50

35

25

(b)

50

75

100 kDa

35

25

TnBglB

81 kDa

Page 225: A THESIS TITLED - prr.hec.gov.pk

Results

195

4.12.2.3. Expression of TnBglA in various Media induced with Lactose

The effect of various cultivation media induced with 150 mM lactose, on the expression of

TnBglA was studied by comparing the heterologous protein profiles of E. coli BL21

CodonPlus (DE3) cultivated in LB, ZB, ZBM, ZYB, 3×ZYB, 4×ZB, M9, ZYBM9, and

3×ZYBM9 inducing media (Figure 4.62).

Figure 4.62: SDS-PAGE analysis of TnBglA (51.5 kDa) in E. coli grown in various lactose inducing media.

Lane M, Protein marker; Lane 1, Control supernatant harboring pET-21a (+) with lactose induction; Lane 2,

Control supernatant harboring pET-21a (+) without induction; Lane 3, Crude supernatant cells proteins

cultivated in ZBM; Lane 4, supernatant cultivated in 3×ZYB; Lane 5, supernatant of ZYB; Lane 6,

supernatant of ZB; Lane 7, supernatant of LB; Lane 8, heat treated supernatant of LB; Lane 9, supernatant of

ZYBM9; Lane 10, heat treated supernatant of ZYBM9; Lane 11, supernatant of 4×ZB; Lane 12, heat treated

supernatant of 4×ZB; Lane 13, supernatant of 3×ZYBM9; Lane 14, heat treated supernatant of 3×ZYBM9.

4.12.2.4. Expression of TnBglB in various Media induced with Lactose

The influence of various cultivation media LB, ZB, ZBM, ZYB, 3×ZYB, 4×ZB, M9,

ZYBM9, and 3×ZYBM9 induced with 150 mM lactose, on the expression of TnBglB was

observed by comparing the heterologous protein profiles of E.coli BL21 CodonPlus (DE3)

cultivated in various inducing media (Figure 4.63).

Page 226: A THESIS TITLED - prr.hec.gov.pk

Results

196

Figure 4.63: SDS-PAGE analysis of TnBglB (81 kDa) in E. coli grown in various lactose inducing media.

Lane M, Protein marker; Lane C, Control supernatant harboring pET-21a (+) without induction; Lane 1,

Crude supernatant cells proteins cultivated in ZBM; Lane 2, heat treated supernatant in ZBM; Lane 3,

supernatant of 3×ZYB; Lane 4, heat treated supernatant of 3×ZYB; Lane 5, supernatant of 3×ZYBM9; Lane

6, heat treated supernatant of 3×ZYBM9; Lane 7, heat treated supernatant of ZB; Lane 8, supernatant of ZB;

Lane 9, supernatant cultivated in 4×ZB; Lane 10, supernatant of 4×ZB; Lane 11, supernatant of LB; Lane

12, heat treated supernatant of LB; Lane 13, supernatant of ZYB; Lane 14, heat treated supernatant of ZYB;

Lane 15, supernatant of ZYBM9; Lane 16, heat treated supernatant of ZYBM9.

4.12.3. Pre-induction optimal cell density

Optimum culture density of both recombinant E. coli BL21 CodonPlus cultures at the time

of induction were determined by using all complex and defined media supplied with both

antibiotics. Recombinant genes was induced at different optical density of culture

(OD600nm) 0.5 to 0.9, and the optimal results were observed when culture was induced at

0.6 optical density in all inducing media, this result was same as described in LB medium.

4.12.4. Effect of Heat Shock

Direct induction of recombinant bacterial host cells at optimal density resulted in

comparatively lower enzymes expression in term of activity as compared to when bacterial

culture was given a heat shock for 60 minutes in a shaking water bath at 42°C with 200 rev

min-1 and after heat shock the culture was induced either with specific concentration of

IPTG or lactose. TnBglA and TnBglB activity were also less when heat shock (42°C) was

kDa

225150100 75 50

35 25

Page 227: A THESIS TITLED - prr.hec.gov.pk

Results

197

given after induction. Similar results were obtained for all inducing media including LB

medium.

4.12.5. Optimal induction temperature, agitation speed

Immediately after heat shock and induction, the bacterial culture was incubated at various

temperatures ranging from 16-40°C in a shaking incubator with different agitation speed

(50-250 rev min-1). Samples were withdrawn periodically from each incubated flasks for

the analysis of both recombinant enzymes activity. The optimal temperature for incubation

was 22°C with an agitation speed of 200 rev min-1, in all studied media. The results were

same as in case of LB medium.

4.12.6. Optimal incubation time

After induction, the incubation time period at which both recombinant culture showed

maximum growth, expression and activity were optimized in all inducing media.

Recombinant enzymes activities and cell growth were monitored up to 96 hours after

regular intervals. TnBglA and TnBglB activity increased up to 72 hours post induction

beyond which further increment in enzymes activities were not detected in any of the media

under study. Therefore, maximum activity was observed after 72 hours of induction at 22°C

in all inducing media (all results were similar with the LB medium finding).

4.12.7. Optimal IPTG concentration

To over-express both heterologous proteins (TnBglA and TnBglB), induced the cloned

culture cells with different concentration of IPTG (0.1-1.0 mM), which revealed that

optimal level of recombinant enzymes activity, expression and bacterial cell density was

observed with 0.5 mM concentration, beyond 0.5 mM, there was no increase in

recombinant genes expression in all used inducing media during fermentation. Moreover,

further increment in IPTG concentration (0.9 mM) retarded the growth of recombinant E.

coli BL21 cells as well as lowered the TnBglA and TnBglB activities (Figure 4.64a and

4.64b). Using IPTG, high cell density (OD600nm) of TnBglA and TnBglB were 16.64 with

9.88 g DCW L-1 and 16.91 with 10.11 g DCW L-1 attained in 4×ZB, respectively which

considerably higher than that observe in LB medium (Table 4.14a and 4.15a).

Page 228: A THESIS TITLED - prr.hec.gov.pk

Results

198

Figure 4.64: Effect of IPTG on recombinant enzymes expression in 4×ZB medium (a) Optimal TnBglA

activity and total cell protein was observed at 0.5 mM IPTG after 72 h incubation at 22°C temperature. (b)

TnBglB showed optimal activity and total cell protein at 0.5 mM IPTG after 72 h incubation at 22°C

temperature, both are defined as 100% activity to calculate the relative activity. Y-error bars show the

standard deviation (±SD) of parallel three experimental reactions, which significant at probability 5%.

4.12.8. Optimal lactose concentration (an alternative inducer)

Recombinant culture production and enzymes expression were studied using lactose a

natural inducer of T7 lac promoter, to scrutinize the effect of lactose various concentrations

(50-300 mM) were used to induce the both cloned TnBglA and TnBglB cultures.

Recombinant TnBglA and TnBglB activities were enhanced gradually with the increment

of lactose concentration up to 150 mM, whereas more increase in concentration appeared

0

1

2

3

4

0

20

40

60

80

100

120

0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 Pro

tein

Co

nc.

(m

g m

L-1

)

Rel

ati

ve

act

ivit

y %

IPTG Concentration (mM)

(a) Optimization of IPTG conc. for TnBglA

IPTG Conc. Protein Conc.

0

1

2

3

4

0

20

40

60

80

100

120

0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 Pro

tein

Con

c. (

mg

mL

-1)

Rel

ativ

e ac

tivi

ty %

IPTG Concentration (mM)

(b) Optimization of IPTG conc. for TnBglB

IPTG Conc. Protein Conc.

Page 229: A THESIS TITLED - prr.hec.gov.pk

Results

199

to be a slightly negative effect or no more increase in activity and expression of enzymes,

approximately in all inducing media (Figure 4.65a and 4.65b). Induction with 150 mM

lactose enhanced the extracellular expression and activity of TnBglA as compared to IPTG

(0.5 mM) induction. While, induction with 150 mM lactose exhibited a same level of

expression in term of activity of TnBglB as in the case of 0.5 mM IPTG.

Figure 4.65: Effect of lactose on the expression of recombinant TnBglA and TnBglB enzymes in 4×ZB

medium (a) Optimal TnBglA activity (expression) and total cell protein was observed at 150 mM lactose

conc. after 72 h incubation at 22°C temperature. (b) TnBglB showed optimal activity and total cell protein at

150 mM lactose conc. after 72 h incubation at 22°C temperature, both are defined as 100% activity to

calculate the relative activity. Y-error bars specify the standard deviation (±SD) of parallel triplicate

experimental reactions. Probability of all the variables is < 5% significance level which entail that elected

variables are significant at 95% confidence interval.

00.511.522.533.544.5

0

20

40

60

80

100

120

50 100 150 200 250 300

Pro

tein

Con

c. (

mg

mL

-1)

Rel

ativ

e a

ctiv

ity

%

Lactose Concentration (mM)

(a) Optimization of Lactose conc. for TnBglA

Lactose Conc. Protein Conc.

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

0

20

40

60

80

100

120

50 100 150 200 250 300

Pro

tein

Con

c. (

mg

mL

-1)

Rel

ativ

e ac

tivi

ty %

Lactose Concentration (mM)

(b) Optimization of Lactose conc. for TnBglB

Lactose Conc. Protein Conc.

Page 230: A THESIS TITLED - prr.hec.gov.pk

Results

200

4.12.9. Comparison of IPTG and Lactose as inducer

Lactose has been found as effective as IPTG for inducing heterologous TnBglA and

TnBglB proteins. Lactose induction, as compared to IPTG, supported exponential growth

of E. coli BL21 CodonPlus (DE3)-RIPL harboring pET-21a–TnbglA and pET-21a–TnbglB

(expression plasmids) to relatively higher cell densities in all inducing media under study

except M9 medium. Maximum optical density (OD600nm) and DCW attained with lactose

inducing medium 18.96 (TnBglA) and 18.66 (TnBglB) which was considerably higher as

compared with 0.5 mM IPTG induction. The culture growth never attained such high-level

optical density as with lactose, even if induced with high level of IPTG concentration,

greater than 0.9 mM IPTG retarded the bacterial growth. Highest cell density (OD600nm) of

E. coli BL21 harboring pET-21a–TnbglA obtained after IPTG induction (0.5 mM), was

16.64 with 9.88 g DCW L-1 in 4×ZB medium, which was noticeably greater than all other

inducing media (Table 4.14a). While, maximum cell density (OD600nm) of recombinant host

cell obtained after lactose induction (150 mM), which was 18.96 with 11.30 g DCW L-1 in

4×ZB medium, significantly greater than all other inducing media (Table 4.14b). TnBglA

enzyme activity was 3.8 fold enhanced in 4×ZB medium when recombinant culture was

induced with 150 mM lactose, and 3.74 fold enhanced in 4×ZB medium with 0.5 mM IPTG

induction as compared to the LB medium.

Optimal cell density (OD600nm) of E. coli BL21 (DE3) harboring pET-21a–TnbglB obtained

after IPTG induction (0.5 mM), was 16.91 with 10.11 g DCW L-1 in 4×ZB medium that

was significantly greater than all other inducing media (Table 4.15a). However, induction

with lactose (150 mM) enhanced the cell density and dry cell weight were 18.66 and 11.08

g DCW L-1 in 4×ZB medium, respectively, which was considerably greater than all other

inducing media used for this recombinant strain (Table 4.15b). In case of TnBglB, lactose

induction (150 mM) exhibited a same level of expression in term of extracellular activity

as in the case of 0.5 mM IPTG; and TnBglB activity was only 0.096 and 0.085 fold

enhanced in 4×ZB medium with lactose and IPTG inducers, respectively as compared to

the LB medium.

Page 231: A THESIS TITLED - prr.hec.gov.pk

Results

201

Table 4.14 (a): Recombinant TnBglA production, induced with 0.5 mM IPTG when cultivated in

different neutral pH media, at 22°C after 72 h induction in a shaker with 200 rev min-1.

Media

0.5 mM IPTG Inducer

Max.

OD600nm

DCW*

(g L-1)

a TCP*

(mg L-1)

Relative activity

(%)

b TnBglA % of

total protein

LB 08.11 04.81 2130 21 22

LB+ 08.19 04.91 2125 19 22

ZB 08.92 05.34 1937 44 22

4×ZB 16.64 09.88 2680 100 30

ZYB 10.16 06.04 2161 61 26

3×ZYB 13.09 07.71 2346 68 26

ZYBM9 08.40 04.98 1917 41 22

3×ZYBM9 09.08 05.38 1987 47 23

ZBM 06.77 04.01 1821 29 21

M9 03.11 01.81 0663 05 11

Table 4.14 (b): Recombinant TnBglA production, induced with 150 mM lactose when cultivated in

different neutral pH media, at 22°C after 72 h induction in a shaker with 200 rev min-1.

Media

150 mM Lactose Inducer

Max.

OD600nm

DCW*

(g L-1)

a TCP*

(mg L-1)

Relative activity

(%)

b TnBglA % of

total protein

LB 09.86 05.86 2116 20 22

LB+ 09.79 05.78 2011 22 22

ZB 10.76 06.39 2086 51 24

4×ZB 18.96 11.30 2865 100 34

ZYB 13.21 07.84 2344 61 32

3×ZYB 14.83 08.81 2463 79 32

ZYBM9 10.45 06.19 2115 47 28

3×ZYBM9 11.16 06.61 2168 63 33

ZBM 10.47 06.20 2294 44 22

M9 03.22 01.88 0756 07 17

*Dry Cell Weight (DCW); *Total Cell Protein (TCP). a Total call protein was estimated by Bradford method.

b Expression levels were computed by densitometric scanning of total cell protein resolved on 12% SDS-PAGE.

Page 232: A THESIS TITLED - prr.hec.gov.pk

Results

202

Table 4.15 (a): Recombinant TnBglB production, induced with 0.5 mM IPTG when cultivated in

different neutral pH media, at 22°C after 72 h induction in a shaker with 200 rev min-1.

Media

0.5 mM IPTG Inducer

Max.

OD600nm

DCW*

(g L-1)

a TCP*

(mg L-1)

Relative activity

(%)

b TnBglB % of

total protein

LB 08.41 05.04 2184 91 28

LB+ 08.29 04.97 2178 82 28

ZB 09.82 05.81 1817 51 18

4×ZB 16.91 10.11 2708 100 30

ZYB 08.16 04.87 2161 69 20

3×ZYB 09.09 05.41 2253 73 21

ZYBM9 11.40 06.84 2417 94 28

3×ZYBM9 07.51 04.48 1754 42 16

ZBM 06.47 03.88 1633 31 16

M9 02.71 01.52 0616 08 09

Table 4.15 (b): Recombinant TnBglB production, induced with 150 mM lactose when cultivated in

different neutral pH media, at 22°C after 72 h induction in a shaker with 200 rev min-1.

Media

150 mM Lactose Inducer

Max.

OD600nm

DCW*

(g L-1)

a TCP*

(mg L-1)

Relative activity

(%)

b TnBglB % of

total protein

LB 09.82 05.78 2183 92 28

LB+ 09.59 05.69 2103 85 28

ZB 10.66 06.29 1935 49 16

4×ZB 18.66 11.08 2817 100 30

ZYB 10.09 06.02 2444 69 20

3×ZYB 11.23 06.66 2574 73 20

ZYBM9 12.37 07.34 2621 86 22

3×ZYBM9 08.26 04.81 2214 43 16

ZBM 07.38 04.34 2188 34 15

M9 03.41 01.96 0684 08 08

*Dry Cell Weight (DCW); *Total Cell Protein (TCP). a Total call protein was estimated by Bradford method.

b Expression levels were computed by densitometric scanning of total cell protein resolved on 12% SDS-PAGE.

Page 233: A THESIS TITLED - prr.hec.gov.pk

Results

203

4.12.10. Purification of Recombinant TnBglA and TnBglB

For the purification of soluble extracellular unrefined TnBglA and TnBglB proteins were

cultivated individually under optimal conditions in fresh sterilized 4×ZB medium (5 L

flasks) supplemented with both antibiotic (as describe earlier). Optimal heterologous

expression and efficient folding of proteins were observed when both neutral pH culture

media were induced at 0.6 OD600nm with lactose (150 mM) after heat shock treatment for 1

h at 42°C followed by incubated for 72 h at 22°C in a rotary shaker (200 rev min-1).

Unrefined cloned TnBglA and TnBglB were purified by employing two subsequent

purification steps of IEC and HIC after heat-treatment (as described earlier). Purity and

activities of enzymes were monitored at each stage of purification by SDS-PAGE analysis,

7.37 fold with a recovery of 28.5% yield and 7.42 fold purification with a recovery of 41%

yield were achieved, respectively. The active fractions of TnBglA and TnBglB exhibited a

single prominent band with 51.50 kDa and 81 kDa on SDS-PAGE, respectively which

revealed that the target heterologous proteins were successfully purified (Figure 4.66a and

4.66b).

Figure 4.66 (a): SDS-PAGE analysis of recombinant purified TnBglA after lactose (150 mM) induction in

4×ZB medium. Lane M, Novagen Protein marker (cat # 69079-3); Lane 1: Purified TnBglA fraction (2 µg)

(b): SDS-PAGE analysis of recombinant purified TnBglB after lactose (150 mM) induction in 4×ZB medium.

Lane M, Novagen Protein marker (cat # 69079-3); Lane 1: Purified TnBglB fraction (2 µg); Lane 2: Purified

TnBglB fraction (2 µg).

75 kDa

100 kDa 81 kDa

(b) (a)

51.5 kDa 50 kDa

35 kDa

150 kDa

225 kDa

25 kDa

Page 234: A THESIS TITLED - prr.hec.gov.pk

CHAPTER-V

DISCUSSION

Page 235: A THESIS TITLED - prr.hec.gov.pk

Discussion

204

5. Discussion

Industrial biotechnology is ubiquitous and has caused a huge impact on diverse industries

especially for green energy generation along with sustainable production of highly valuable

genetically engineered products as compared to previously envisioned (Elleuche et al.,

2014). Recently, biotechnological products have gained significant interest all over the

world, which are proficient of enhancing the efficiency of bioenergy production, livestock,

agriculture, pharmacy, poultry, paper and pulp, beverage, food, detergent and textile

industries. The prevailing conditions in industrial processes are frequently far from the

properties of standard biocatalysts. Therefore, the demand of novel highly stable and

efficient biocatalysts is considerably increased globally, which are capable to reach the

goal (aim) by supplementing or replacing the traditional chemical approaches (Woodley,

2013).

Naturally, the members of genus Thermotoga have been reported to be a valuable home of

highly active and thermotolerant glycoside hydrolases (GHs) especially cellulolytic

enzymes (Mehmood et al., 2014). The highly heat stable cellulases from genus Thermotoga

are the most prominent candidates, which can perform their catalytic functions effectively

under extremely harsh conditions of biosaccharification process and numerous other

industries (Xie et al., 2015; Long et al., 2016). The complete genome (1809823 bp)

sequence of T. naphthophila RKU-10T (GenBank Accession No. CP001839.1) is available

on the NCBI online database. Two putative cellulolytic β-1,4-glucosidases genes TnbglA

(ADA66698.1) and TnbglB (ADA66752.1) from a hyperthermophilic anaerobic bacterium

T. naphthophila were selected for cloning and overexpressed in mesophilic host E. coli

BL21 CodonPlus (DE3)-RIPL using pET-21a(+) expression vector followed by respective

heterologous enzymes purified to homogeneity.

Initially, the amplicons of TnbglA (1.341 kb) and TnbglB (2.169 kb) genes were ligated

into pTZ57R/T vector for direct cloning and sequencing, TA cloning is one of the simplest

and highly proficient strategy to clone genes (Zafar et al., 2014; Haq et al., 2015b). During

primer designing, NdeI restriction site was introduced in forward primer at 5' end, for the

directional cloning and to make TnbglA and TnbglB genes compatible with open reading

Page 236: A THESIS TITLED - prr.hec.gov.pk

Discussion

205

frame (ORF) of pET-expression vector, similar strategy was observed in many earlier

studies (Yang et al., 2015; Chan et al., 2016; Long et al., 2016). The recombinant pTZ-

TnbglA and pTZ-TnbglA plasmids were propagated in E. coli DH5α to maintain a high

copy number, subsequently positive transformants selected by blue white screening, and

positive clones of both genes (TnbglA and TnbglB) were subjected to sequencing.

GHs have been classified on the basis of nucleotide and amino acid sequence similarities,

in which β-glucosidases belong to either GH family 1 or GH family 3 (Liu et al., 2012). T.

naphthophila possesses two different β-glucosidases (TnbglA and TnbglB) genes. Many

thermophiles contain genes for two or more exo-acting β-specific glycosyl hydrolases,

including T. neapolitana (Turner et al., 2007), T. maritima (Xue et al., 2009), T. petrophila

(Haq et al., 2012b), Thermoanaerobacterium thermosaccharolyticum (Pei et al., 2012) and

T. petrophila (Xie et al., 2015). TnBglA and TnBglB displayed high amino acid sequence

homology range of 99-55% with the β-glucosidases of other microbes (thermophiles to

mesophiles). The deduced sequence homology of both β-glucosidases from T.

naphthophila revealed that TnBglA should be classified as the member of GH family 1

because it shared similar consensus sequence blocks (conserved region) with other β-

glucosidases of GH family 1 (Figure 4.11a); while, TnBglB should be classified as the

member of GH family 3 since it shared similar consensus sequence blocks (conserved

region) with other GH family 3 β-glucosidases (Figure 4.11b).

Sequence multi-alignments studies revealed that TnBglA has some conserved regions and

shared highest identity with other GH family 1 thermophilic β-glucosidases especially with

the members of genus Thermotoga. Mostly, conserved regions of TnBglA located near N

and C terminal, there were some conserved regions in which first region YQIEG from 19

to 23, second consensus sequence YRFS from position 76 to 79, third YHWDLP from 120

to 125, fourth NEPW from 165 to 168, fifth TENGAA from 350 to 355, and sixth

DNFEWA consensus sequence from 402 to 407 amino acid residues. In conserved regions,

there were conserved sequences of glycine (G), histidine (H), glutamic acid (E), asparagine

(N), serine (S), tryptophan (W) and phenylalanine (F); and for GH family 1 β-glucosidases,

two strictly conserved catalytic residues of glutamic acid (E), one in N-terminal and the

other in middle or C-terminal region, participate in the hydrolysis of glycosidic bond

Page 237: A THESIS TITLED - prr.hec.gov.pk

Discussion

206

(Jenkins et al., 1995), by a general acid/base catalytic retaining mechanism. TnBglA

protein has conserved catalytic residues, one glutamic acid (E166) act as proton donor closer

to the N terminus and other two glutamic acid (E351 and E405) act as nucleophiles (Figure

4.11a) (Gräbnitz et al., 1991).

The catalytic active sites for GH family 1 β-glucosidases have been identified and

confirmed by site-directed mutagenesis (Wang et al., 1995; Moracci et al., 1996;

Vallmitjana et al., 2001). TnBglA belonged to GH family 1, which shares a retaining

catalytic mechanism by involving three active glutamic acid (E166, E351 and E405) residues.

However, Serine (S) also assists in the hydrolytic activity of β-glucosidase while Gly (G)

provides the turns which are essential and indispensable for the structural stability. The

sequence alignment revealed that the asparagine (N162) residue from TnBglA, is highly

conserved in clan A of GHs except GH family 26, which suggests that it provide a polar

interaction with glucose (Gloster and Davies, 2010). The study of conserved domains of

TnBglA using NCBI database explained that TnBglA is a monomeric protein (Marchler-

Bauer et al., 2015). Previously, a cloned monomeric GH family 1 β-glucosidase from

Thermotoga petrophila has been reported to contain three similar catalytic conserved

glutamic acid (Glu) residues (Haq et al., 2012b). Mostly, GH family 1 thermophilic

monomeric recombinant β-glucosidases have two comparable conserved glutamic acid

(Glu), which suggested as a nucleophile and catalytic acid/base residue (Fang et al., 2014;

Rajoka et al., 2015; Yang et al., 2015; Chan et al., 2016).

Multi-alignments studies of TnBglB with some other GH family 3 members, demonstrated

that it has some highly conserved regions and exhibited great sequence similarity with β-

glucosidases of other Thermotoga species (Goyal et al., 2001; Turner et al., 2007; Xie et

al., 2015). The conserved motifs of TnBglB was observed ADGPAG (57-62), TAFPV (79-

83), GRNFEYYSEDP (129-139), GVQS (152-155), NNQETNR (168-174), LREIYL

(186-191), YNKLNG (210-215), GFVMSDW (237-243), RISGEG (454-459),

NPSGKLPTTFP (537-547), IYVGYRYYDTF (576-586) and RVGASSRDI (697-705)

residues. In conserved regions, there were conserved sequences of glycine (G), lysine (K),

aspartic acid (D), glutamic acid (E), arginine (R), tryptophan (W), threonine (T), leucine

(L), serine (S), tyrosine (Y) and histidine (H) (Figure 4.11b). The sequence alignment of

Page 238: A THESIS TITLED - prr.hec.gov.pk

Discussion

207

TnBglB with other β-glucosidases of GH family 3, showed that one catalytic residue

aspartic acid (D242) present in highly conserved region of domain-I, act as a fully conserved

catalytic nucleophile; and previously this residue has been predicted as active nucleophile

over 15 different GH family 3 β-glucosidases after sequence alignment (Dan et al., 2000;

Turner et al., 2007). Such conservation is consistent with a key catalytic nucleophilic role

in forming the glycosyl-enzyme intermediate by covalent linkage, stabilizing and

modulating the ionization state of proton donor catalytic residue and making a hydrogen

bond to sugar 2-hydroxy group at transition state (Parry et al., 2001). Another general

acid/base catalytic residue glutamic acid (E458) present in highly conserved region of

domain-II, which act as proton donor and display hydrolytic function to break glycosidic

bond. However, the importance of catalytic nucleophile (D242) and essential acid/base

residue (E458) have been demonstrated and confirmed by site-directed mutagenesis (or

selected active-site variants) at the catalytic pocket (Hong et al., 2006; Pozzo et al., 2010;

Zhao et al., 2015).

The signature amino acid sequence analysis revealed that TnBglB consists of a three-

modular domain protein, from the N-terminal to C-terminal, having glycosyl hydrolase

family 3 N-terminal catalytic domain (Glyco_hydro_3), a glycosyl hydrolase family 3 C-

terminal catalytic domain (Glyco_hydro_3_C), and a fibronectin type III (FnIII) module

found at C-terminal end, its actual function is unknown although FnIII domain is closely

associated with N- and C-terminal glycosyl hydrolase domains (Marchler-Bauer et al.,

2015). The conserved sequence KHFV (163 to 166) of TnBglB is a putative carbohydrate-

binding site (Thongpoo et al., 2013). GH family 3 β-glucosidases from Thermotoga

neapolitana (Pozzo et al., 2010), T. petrophila (Xie et al., 2015) and Myceliophthora

thermophila (Zhao et al., 2015) also contained three domains with similar pattern. All the

data presented here reveal that TnBglB follows a retention catalytic mechanism, and a

prominent member of GH family 3 composed of three domains with two highly active

residues, D242 and E458 in catalytic center.

Both genes TnbglA and TnbglA were sub-cloned in an efficient expression vector pET-

21a(+) under the control of a T7 lac promoter and the strong upstream translation signals

for target proteins, followed by optimally overexpressed in a most popular, well-

Page 239: A THESIS TITLED - prr.hec.gov.pk

Discussion

208

established and excellent expression platform E. coli BL21 Codon Plus (DE3)-RIPL host

strain. For overexpression of thermostable β-glucosidases genes various expression vectors

and mesophilic hosts were efficaciously used, whereas the pET expression vector and E.

coli BL21 CodonPlus (RIPL) host is the most preferable expression coordination as

compared to others. Due to high growth rate, well characterized molecular genetics and

incredible expression level, many researcher have been selected pET vector and E. coli

BL21 CodonPlus strain to overexpressed β-glucosidases isolated from Dictyoglomus

thermophilum (Zou et al., 2012), Thermotoga thermarum (Long et al., 2016) and

Caldicellulosiruptor owensensis (Peng et al., 2016).

Positive transformants of E. coli BL21 harboring the pET-21a–TnbglA and pET-21a–

TnbglB plasmids were cultivated initially in LB medium and induced with IPTG, subjected

to SDS-PAGE for the analysis of heterologous expression of proteins along with crude

control supernatant of E. coli BL21 harboring pET-21a (+) without insert, which also

induced with IPTG. The crude media supernatant of TnBglA and TnBglB enzymes

displayed a prominent band of 51.5 kDa and 81.1 kDa on SDS-PAGE, respectively (Figure

4.23a and 4.23b). Both cloned enzymes from T. naphthophila did not have putative signal

peptide sequence; expression and activity were observed in all three cellular fractions but

extracellular soluble fraction exhibited maximum expression as compared to cell-free

lysate and cell-bound fractions (Figure 4.27a and 4.27b). Similar behavior of cloned

thermophilic β-glucosidases have been reported from Thermoascus aurantiacus (Parry et

al., 2001).

To attain a high level expression of recombinant enzymes and cultures density, a better

cultivation approach has developed by modifying and optimizing a few essential

parameters such as pre-induction optical cell density, growth temperature, better agitation,

heat shock treatment (42°C for 1 h) to the bacterial cultures before and after induction, and

optimal inducer concentration which played a critical role to improve the bacterial growth

and revamp the targeted proteins with proper folding. Best pre-induction optical density

(OD600nm) of TnBglA and TnBglB cultures for getting maximum expression were studied.

High level of expression and total cell proteins were observed when both cultures induced

at 0.6 (OD600nm) (Figure 4.29a and 4.29b), which indicated that host cell density effects the

Page 240: A THESIS TITLED - prr.hec.gov.pk

Discussion

209

output of heterologous proteins, similar behavior had been reported in many earlier studies

(Schroder et al., 2014; Yang et al., 2015; Chan et al., 2016).

Effect of heat shock treatment on the expression of both enzymes were reconnoitered,

TnBglA and TnBglB expression and activities were greatly improved when cultures were

given heat shock at 42°C for 1 h just before induction (Figure 4.30a and 4.30b). Heat shock

treatment to the bacterial culture is accountable for the production of molecular chaperons

(heat shock protein), which are concerned primarily with protein folding and support the

appropriate expression, and constrain the formation of inclusion bodies (Oganesyan et al.,

2007; Hameed et al., 2014). After heat shock treatment, both cultures were induced with

different concentration of IPTG (0.1-0.9 mM), and optimal expression of TnBglA and

TnBglB were observed with 0.5 mM IPTG (Figure 4.31 and 4.32). Cell density and

enzymes expression were reduced with the further increment of IPTG molar concentration

because high amount of IPTG causes a toxic effect on host cells growth. Use of reduced

concentration of IPTG is an effective approach to get the high level of soluble protein

expression and host cell growth, which also make the fermentation process more

economical. Similar strategy has been applied by many researchers to achieve better

expression of β-glucosidases such as Kim et al. (2012), Schroder et al. (2014), Xie et al.

(2015), and Chan et al. (2016).

Inducing temperature is a critical parameter, as can be observed its effect on the

heterologous proteins expression and host cell growth. Therefore, effect of induction

temperature (16-42°C) on the expression of TnBglA and TnBglB were investigated by

incubating culture at various temperature after induction with IPTG (0.5 mM). Best

expression and production outputs of recombinant enzymes were obtained when culture

incubated at 22°C in a shaking incubator (Figure 4.33a and 4.33b). Low temperature

inducement is an effective method for proper protein folding and to achieve maximum

soluble expression. This strategy is also effectual to repress the production of inclusion

bodies. Several group of researchers have been reported that optimal expression of β-

glucosidases were observed at low temperature inducement as compared to 37°C and

reduced the inclusion bodies (Xue et al., 2009; An et al., 2010; Xie et al., 2015).

Page 241: A THESIS TITLED - prr.hec.gov.pk

Discussion

210

Time of incubation, pH of medium and agitation speed are another important factors which

affect the production and expression of desired protein along with growth of engineered E.

coli CodonPlus. Therefore, time course profile up to 96 hours was studied in various pH

(5.5-8.0) of LB medium along with different agitation speed (50-250 rev min-1) after

induction. Optimal expression of both heterologous proteins were observed in pH 7.0

medium after 72 h inducement at 22°C with 200 rev min-1. Previously, a study

demonstrated that best expression of β-glucosidase was achieved when E. coli BL21 host

cells grown at 18°C for 20 hours in a shaking incubator with 200 rev min-1 after IPTG

induction (Wang et al., 2012). Hence, low temperature along with appropriate

environmental conditions and inducement time are necessary factors for the proper

recombinant protein folding into native conformation.

For purification of TnBglA and TnBglB, host cells grown individually at optimal

cultivation conditions followed by stockpiled culture supernatants. Partial purification of

both enzymes were achieved by heat treatment at 70°C for 1 h. At such high temperature

approximately all heat-liable proteins of mesophilic host were denatured and thermo-

tolerant cloned protein was partially purified. SDS-PAGE analysis revealed that heat-

precipitation step was alone sufficient to remove several host contaminating proteins

(Figure 4.38a and 4.38b). Heat denaturation is an inexpensive, highly effective and an

expedient prime enrichment step in refining of thermostable recombinant enzymes

expressed in mesophilic host. Formerly, several thermophilic bacterial β-glucosidases

partially purified using this strategy (Li et al., 2013; Xie et al., 2015; Gumerov et al., 2015;

Long et al., 2016).

TnBglA and TnBglB were further purified to homogeneity using anion-exchange (AEC)

and hydrophobic interaction (HIC) chromatography, activity of both enzymes were

monitored at each step of purification (Table 4.7a and 4.7b), similar scheme of purification

has been adopted by Haq et al. (2012b). TnBglA and TnBglB were purified with a final

purification of 7.28 and 7.38-fold, a yield of 40.8 and 39.5% with the highest specific

activity of 255×103 U mg-1 and 130×103 U mg-1, respectively. Purification folds are largely

consistent with the results obtained by Shin and Oh, (2014). The apparent molecular mass

and purity of TnBglA and TnBglB enzymes were analyzed by SDS-PAGE, which showed

Page 242: A THESIS TITLED - prr.hec.gov.pk

Discussion

211

a single prominent band of 51.50 kDa and 81.1 kDa, respectively (Figure 4.38a and 4.38b).

Molecular mass of purified TnBglA is in agreement with those of many GH family 1 β-

glucosidases characterized from other thermophilic bacteria (Pei et al., 2012; Zou et al.,

2012; Yang et al., 2015). Purified TnBglB has a molecular mass of 81.1 kDa which is in

accordance to many other GH family 3 β-glucosidases characterized from other

thermophilic bacterial sources (Goyal et al., 2001; Turner et al., 2007; Xie et al., 2015).

Extracellular purified TnBglA displayed a single noticeable band on native-PAGE under

non-reduced conditions and after boiling (Figure 4.39a), which suggests that TnBglA is a

monomeric protein. This behavior is similar to β-glucosidases as from Thermotoga

neapolitana (Park et al., 2005) and T. thermarum (Long et al., 2016). While, under non-

denatured conditions and after boiling, TnBglB enzyme remained as a trimer with high

molecular mass monomeric conformation and showed a single band on native-PAGE

(Figure 4.39b). Similarly, Parry et al. (2001) had been reported a trimeric GH family 3 β-

glucosidase from Thermoascus auranticus with a 120 kDa monomer under non-reducing

conditions.

Individually, purified TnBglA and TnBglB were completely characterized; variation of

temperature and pH obviously influenced the activity of enzymes. To reconnoiter pH

dependence of both enzyme activity, employed an overlapping buffer systems to cover a

broad range of pH from 3.0 to 11.0 at 70°C, and also investigate the optimal buffer system

for the catalytic efficacy. Generally, different buffer system greatly influenced the enzymes

activities, hence McIlvaine buffer for pH 3.0-7.0, HEPES for pH 8.0 and for pH 9.0-11.0

CAPS were found the best buffer for both enzymes. Peak hydrolytic activity of TnBglA

and TnBglB were determined at pH 7.0 and 5.0, respectively using McIlvaine buffer, and

a noteworthy difference was observed in activity between McIlvaine and Tris-Cl buffer at

optimal pH of both enzymes. Maximum activity in McIlvaine buffer as compared to other

buffer systems, thought to be due to charge distribution or changes in conformation.

Similarly, reduction in catalytic activity of β-glucosidases in Tris-Cl and optimal in

McIlvaine buffer had been reported from Exiguobacterium sp. DAU5 (Chang et al., 2011)

and Sphingopyxis alaskensis (Shin and Oh, 2014).

Page 243: A THESIS TITLED - prr.hec.gov.pk

Discussion

212

Purified TnBglA retained more than 90% of optimal activity at 6.0 to 9.0 broad range of

pH, and enzyme efficiency was completely repressed at high concentration of hydrogen

ions and more alkaline aqueous environment (Figure 4.40a). Whereas, purified TnBglB

exhibited 90% catalytic activity at pH 4.0 and reduced in alkaline pH range (Figure 4.40b).

Optimal pH of both enzymes are not unexpected, as most bacterial β-glucosidases showed

maximum activity at neutral or slightly acidic ranges (Fang et al., 2014). Any change in

ionic strength from optimal pH caused an obvious constrain on catalytic efficiency because

enzymes are extremely sensitive to change in pH that can alter structural conformation by

dissolving hydrogen and ionic bonds, which subsequently denature the catalytic site and

eventually disrupt the enzyme-substrate complex (Stoker, 2008; Ha and Bhagavan, 2011).

Comparison of the optimum pH of TnBglA with other thermophilic bacterial GH family 1

β-glucosidases revealed that it was quite similar to Dictyoglomus thermophilum (Zou et

al., 2012) and T. maritium (Mehmood et al., 2014); higher than the previously reported β-

glucosidases from Thermoanaerobacterium aotearoense (Yang et al., 2015), and

Acidilobus saccharovorans (Gumerov et al., 2015) that displayed peak activity at pH 6.0.

Although, some variant β-glucosidases from T. thermarum (Zhao et al., 2013) and

Anoxybacillus sp. (Chan et al., 2016) optimally active at pH 4.8 and 8.5, respectively.

Optimal pH of TnBglB is fairly similar to other bacterial GH3 β-glucosidases from T.

petrophila (Xie et al., 2015) and T. thermarum (Long et al., 2016). However, it was higher

than that of previously reported GH3 β-glucosidases from Thermofilum pendens (Li et al.,

2013) that displayed maximum activity at pH 3.5, and lower than from Terrabacter

ginosenosidimutans (An et al., 2010) that exhibited peak activity at pH 7.0.

Purified TnBglA and TnBglB displayed peak activity at 95 and 85°C, respectively. TnBglA

retained more than 90% catalytic activity at 100°C (Figure 4.41a), while TnBglB showed

more than 85% activity even at 90°C, and at 100°C TnBglB activity declined up to 66%

(Figure 4.41b). At optimal temperature, catalytic reaction rate become faster due to high

kinetic energy between the enzyme-substrate complexes. Whereas, enzyme denature

rapidly above the optimum temperature, due to the disruption of approximately all

hydrogen bonds and weak-interactions, which are accountable for the stability and proper

folding of enzyme catalytic site (Laidler and Peterman, 2009; Daniel et al., 2010).

Page 244: A THESIS TITLED - prr.hec.gov.pk

Discussion

213

Optimal temperature of TnBglA (95°C) is analogous to the thermophilic recombinant GH1

β-glucosidases from T. maritima (Mehmood et al., 2014) and Acidilobus saccharovorans

(Gumerov et al., 2015); and higher than those reported from Thermoanaerobacterium

aotearoense (Yang et al., 2015) and Anoxybacillus sp. (Chan et al., 2016) that optimally

work at 60°C and 70°C, respectively. However, a β-glucosidase isolated from a

hyperthermophilic Pyrococcus furiosus archaeon had an exceptional 105°C optimal

temperature (Voorhorst et al., 1995). TnBglB optimal temperature (85°C) is similar to other

GH3 β-glucosidases cloned from Dictyoglomus turgidum (Kim et al., 2011) and T.

thermarum (Long et al., 2016). While, higher than β-glucosidases from Myceliophthora

thermophila (Zhao et al., 2015) and lower than from T. petrophila (Xie et al., 2015).

Optimal incubation time for the substrate hydrolysis is a crucial parameter. Therefore, time

course profile for the hydrolysis of substrate using purified TnBglA and TnBglB were

studied from 01 to 15 minutes. Both enzymes showed peak hydrolytic activity after 10

minutes incubation of enzyme-substrate mixture however further increase in incubation

time did not cause any perceptible effect (Figure 4.42). Optimal incubation time of TnBglA

and TnBglB is in accord with previously reported cloned GH1 β-glucosidases (Hong et al.,

2009; Fang et al., 2014; Schroder et al., 2014), and GH3 β-glucosidases (Goyal et al., 2001;

Zhao et al., 2015; Long et al., 2016).

Thermostability is the most prominent feature of β-glucosidases that cloned from

hyperthermophiles especially from the members of genus Thermotoga, which make them

an imperative candidates of several biotechnological and industrial applications. Thermal

stability profile revealed that TnBglA and TnBglB were thoroughly stable at 85°C for 720

minutes (12 h) and 480 minutes (8 h), respectively at neutral pH range from 6.5 to 7.5

(Figure 4.43a and 4.43b), while lowest thermostability of both enzymes were found at pH

4.0, which suggesting that the best ionization state and net charge for stabilization of

enzyme structure maintains at narrow range of pH (6.5 to 7.5), which is different from the

optimum net charge of TnBglB for catalysis. Net charge acquired at neutral pH is sufficient

to stabilize the secondary and tertiary structure of recombinant TnBglA and TnBglB even

at higher temperature (Gitlin et al., 2006; Colussi et al., 2015). Conversely, above 80°C

and at low pH 4.0, alternation and structural destabilization occur at both secondary and

Page 245: A THESIS TITLED - prr.hec.gov.pk

Discussion

214

tertiary level, and net charge can influence their equilibrium properties such as free energy

of enzyme interactions with ligands.

Purified TnBglA showed astonishing thermostability as compare to TnBglB, at 90°C,

TnBglA retained 82% residual activity after being incubated for 300 minutes (5 h) at pH

6.5 to 7.5, whereas TnBglB lost 46% of its original hydrolytic efficiency after 240 minutes

(4 h) incubation. Thus, hyperthermostable TnBglA and TnBglB could be used efficaciously

in various fields such as in food processing and saccharification of biomass, etc. The

variant thermotolerant behavior of both enzymes with different pH level, is in harmony

with previously reported cloned β-glucosidases from T. petrophila (TpBgl1 from GH1 and

TpBgl3 from GH3) and Pyrococcus furiosus (PfBgl1 from GH3) that showed highest

thermostability at pH 6.0 and lowest at pH 4.0 (Cota et al., 2015).

Thermal stability of TnBglA was higher than the stability of other GH1 β-glucosidases

from Fervidobacterium islandicum (Jabbour et al., 2012) and T. thermarum (Zhao et al.,

2013) that displayed 100% thermal efficiency at 70°C for 3 h and 85°C for 2 h, respectively

at neutral pH. While, an exceptional β-glucosidase from T. maritima was fairly stable up

to 140°C (Mehmood et al., 2014). Thermal tolerance of TnBglB is greater than the several

GH3 β-glucosidases from Dictyoglomus turgidum (Kim et al., 2011) and T. thermarum

(Long et al., 2016) that remained active at pH 5.0 up to 85°C and 75°C for 5.57 h and 2 h,

respectively. But an exceptional acidophilic and thermo-active GH3 β-glucosidase from

Thermofilum pendens maintained nearly 100% of its peak activity at pH 4.0 after 2 h

incubation at 90°C (Li et al., 2013).

Both purified TnBglA and TnBglB displayed an astounding pH stability, which indicated

their potential as industrial candidates. Bu et al. (2013) suggested that enzymes catalytic

activities are directly linked with the balance between protonation and conformational

changes. In this present study, TnBglA remained stable over a broad range of pH from 6.0

to 9.0, with a drastic reduction to ≈3% at pH 3.0 (Figure 4.44a). While, TnBglB remained

stable at pH 6.5 to 7.5 and retained more than 70% of its optimal activity at pH 5.0 to 8.5

(Figure 4.44b). Hence, the experimental changes in the hydrolytic behavior of TnBglA and

TnBglB with variation in pH were due to modified protonation of enzyme’s amino acid

Page 246: A THESIS TITLED - prr.hec.gov.pk

Discussion

215

residues and net charge that are directly affecting the interaction with ligands. In enzyme

catalysis, ionic groups are involved, for instance, acid-base amino acid residues in the

active site of β-glucosidases (Colussi et al., 2015). Therefore, the protonation state of the

carboxylate nucleophile and carboxylic acid amino acid residue are essential for enzymatic

reaction and a variation in pH could impair the hydrolytic mechanism (Gitlin et al., 2006;

Sørensen et al., 2013).

The result of TnBglA stability is in agreement to the numerous formerly reported GH1 β-

glucosidases from Lactococcus sp. FSJ4 (Fang et al., 2014), and Anoxybacillus sp. (Chan

et al., 2016) which showed stability within a broad range of pH. While, TnBglB showed

more pH stability at neutral range than the acidic and alkaline conditions, this phenomenon

is in accordance to other cloned GH3 β-glucosidases from T. petrophila (Xie et al., 2015)

and T. thermarum (Long et al., 2016). However, in contrast to TnBglB, an acidophilic β-

glucosidase from Thermofilum pendens retained 80% of its maximum activity at an acidic

range of pH 3.5 to 4.0 (Li et al., 2013).

Recombinant TnBglA and TnBglB enzymes exhibited remarkable thermal and pH stability,

can retained their catalytic activity for a longer time and reduce amount will need in the

reaction processes due to reusability. Steam explosion is commonly applied during the

biosaccharification process to make plant biomass more accessible and suitable for

dynamic cellulolytic hydrolysis, a highly thermostable β-glucosidase (either TnBglA and

TnBglB) could be used simultaneously along with other thermo-efficient cellulases

(endoglucanases and cellobiohydrolases) and directly devoid of a pre-cooling step thereby

can reduce the processing time and cost, save energy, provide an opportunity to increase

solubility of products and reactants, decrease the risk of contamination, improve qualities

and yields (Pei et al., 2012). In practical applications, thermal activation is desired since

longer active life means the less enzyme consumption, due to the incredible stability of

TnBglA and TnBglB at elevated temperature and pH, both are the potential candidates for

wide industrial bioconversion processes, including waste treatment, textiles, paper and

pulp for cellulose-degrading purpose (Kuhad et al., 2011).

Page 247: A THESIS TITLED - prr.hec.gov.pk

Discussion

216

The great storage stability of crude supernatant, heat treated (at 70°C for 1 h) and purified

TnBglA and TnBglB were observed after storage at different conditions like room

temperature and 4°C, as shelf-life of an enzyme is an imperative parameter for the

commercial utilization. Providentially, enzymes exhibited an excellent storage stability and

retained 100% of original activity after storage at room temperature for about 175 days

while crude supernatant showed marginal decline in activity of 5-7%, and no substantial

loss of activities were observed at 4°C (up till now) in all the three ways of storage (crude

supernatant, heat treated and purified). Schroder et al. (2014) reported that a heat-active β-

glucosidase from hydrothermal spring was absolutely stable at 4°C. In contrast to the

present study, a cellulolytic enzyme from Trichoderma viridae had lost its original activity

up to 42% after 30 days storage at room temperature (Iqbal et al., 2011).

Usually chaotropic agents, trace metals and detergents modify the original activity of

biocatalyst to a certain limit or may terminate the catalytic efficiency completely.

Therefore, inhibitory effect of some chemical agents and metallic cations on both enzymes

were assessed. Divalent metallic cations (Ca2+, Cd2+, Co2+, Fe3+, Mg2+, Mn2+, Ni2+, Zn2+,

Pb2+ and Cu2+), chelating agent (EDTA), detergents (Triton X-100 and Tween-80) and urea

did not exhibit any sensitive influence on TnBglA and TnBglB catalytic efficiency (Figure

4.45a and 4.45b), which revealed that these additives were not necessary for the enzymes.

The results also explains that catalytic region of both proteins do not have a potential metal

ion binding site. Therefore, TnBglA and TnBglB are determined as metal-independent

proteins. However, their activities were intensely inhibited by Hg2+ and SDS which proved

to be the strong inhibitors; and suggested that the active site might have thiol groups which

involved in binding or catalysis and necessary for the proper and accurate tertiary structure

of enzyme (Liu et al., 2012).

Distinctly, TnBglA displayed the most favorable property that is the independence of metal

ions and various chemical additives. Largely congruent with the other reported bacterial

GH1 β-glucosidases as from T. thermarum (Zhao et al., 2013) and Anoxybacillus sp. (Chan

et al., 2016) recognized to be various additives tolerant. In contrast, hydrolytic activity of

multiple GH1 β-glucosidases modified to some extent or completely repressed by the

addition of these agents as have been explained in various studies (Chang et al., 2011; Lu

Page 248: A THESIS TITLED - prr.hec.gov.pk

Discussion

217

et al., 2013; Yang et al., 2015). Conspicuously, TnBglB affinity was slightly improved by

the addition of Ca2+, Zn2+, Mn2+ and Co2+ cation by 125%, 120%, 115% and 110%,

respectively. These cations play a significant function in structural stability and improve

the binding affinity of substrate by stabilizing the active site conformation. Hence, it is

likely that β-glucosidase may has a metal ion binding loop, which contains the competency

to retain protein structure by binding with ions. An increased activity with metal ions and

repressed by Hg2+, were recently described for GH3 thermophilic β-glucosidases from

Thermofilum pendens (Li et al., 2013) and T. petrophila (Xie et al., 2015). TnBglB activity

was significantly reduced by SDS, but no obvious influence was detected with the addition

of EDTA (Long et al., 2016).

In addition, disulfide reducing agent β-mercaptoethanol enhanced the activity of TnBglA

and TnBglB up to 110% and 146%, respectively (Figure 4.45a and 4.45b). It is a well-

known inhibitor of thiol group, suggesting that sulfhydryl group (SH-group) may not be

engaged in catalytic center of both enzymes structure, while rather may be necessary for

maintaining three-dimensional structure and also play role in sustaining the enzyme in

reducing state. There is, although rare, precedent for the activation by β-mercaptoethanol

has been observed in β-glucosidases from T. neapolitana (Park et al., 2005) and

hydrothermal spring metagenome (Schroler et al., 2014); while Jabbour et al. (2012)

reported that enzyme activity was repressed by β-mercaptoethanol.

Mostly, organic solvents like acetone, ethyl acetate, acetonitrile, dimethyl sulfoxide

(DMSO) and dimethylformamide (DMF), and straight chain aliphatic alcohols (methanol,

ethanol, n-butanol and iso-propanol) caused strong inhibitory influence. TnBglA tolerated

competently a variety of organic solvents at low concentration, and no obvious effect has

been perceived with 15% (v/v) organic solvents though at high concentration the activity

decreased drastically (Figure 4.46a). This demeanor is largely congruent with the results

obtained by other GH1 β-glucosidases, where all these organic solvents revealed an

inhibitory effect on enzymes (Jabbour et al., 2012; Schroder et al., 2014). Moreover, Fang

et al. (2010) has also been reported that β-glucosidases lost its original activity to 30% with

5% (v/v) DMSO. In this study, purified TnBglB showed more resistance attitude towards

organic solvents as compared to TnBglA, and acts as a competitive inhibitors. No

Page 249: A THESIS TITLED - prr.hec.gov.pk

Discussion

218

significant influence was found with 2-25% (v/v) organic solvents and caused little to

moderate reduction in TnBglB activity with the increase concentration (Figure 4.46b). This

agree with the data for GH3 β-glucosidase from T. maritima (Goyal et al., 2001); while

some other β-glucosidases also displayed similar reduction behavior with the increment of

DSMO (Li et al., 2013; Xie et al., 2015).

However, TnBglA was unaffected at low concentration of methanol, ethanol, n-butanol and

iso-propanol, but drastically reduced up to 8-10% in the presence of 30% (v/v) alcohols

(Figure 4.47a). This trend of reduction in activity by alcohols were in agreement with the

result of Schroder et al. (2014). Although rare, there is precedence in the enhancement of

GH1 β-glucosidase activity as has been reported by Zhao et al. (2013). TnBglB was not

highly influenced by straight chain aliphatic alcohols, although low concentration

considerably activated the enzyme followed by suppressed up to certain limit with 50%

(v/v) (Figure 4.47b). This result demonstrated that TnBglB can be used effectively in

organic biotransformation, and the stimulatory effect of alcohols is largely attributed to the

occurrence of transglycosylation or to allosteric interactions. Similarly, positive influence

by alcohol on recombinant GH3 β-glucosidases have been reported from Thermofilum

pendens (Li et al., 2013) and T. petrophila (Xie et al., 2015).

Cellobiose is a potent competitive inhibitor of endoglucanases and cellobiohydrolases, β-

glucosidase is commonly a rate-limiting factor during hydrolysis and highly sensitive to

glucose inhibition. Hence, a β-glucosidase with strong resistance to the end product

(glucose) inhibition is an attractive candidate in this field. By studying the effect of glucose

(20-1500 mM), it was revealed that TnBglA not only resistant to glucose inhibition, on the

contrary its activity is perceptibly enhanced by glucose (20-600 mM), and displayed the

high apparent affinity constant or inhibition constant (Ki) value of 1200 mM (Figure 4.48a).

Mostly, bacterial β-glucosidases did not exhibit resistance to glucose (end product),

however rare displayed some tolerance to glucose. Ki value of TnBglA for glucose

inhibition is higher as compared to many glucose-tolerant GH1 β-glucosidases (Cota et al.,

2015; Gumerov et al., 2015; Yang et al., 2015).

Page 250: A THESIS TITLED - prr.hec.gov.pk

Discussion

219

Various concentration of glucose (20-600 mM) was used to investigate the effect of glucose

on TnBglB activity and determined the Ki value for glucose inhibition. Recombinant GH3

TnBglB exhibited less glucose tolerant behavior as compared to GH1 TnBglA, and the

activity of TnBglB decreased rapidly at high concentration of glucose. It has been observed

frequently that β-glucosidases belonging to GH family 3 showed less resistant to glucose

than family 1. However, TnBglB has a Ki value of 150 mM (Figure 4.48a), which is higher

than the previously reported recombinant bacterial GH3 β-glucosidase isolated from T.

petrophila with Ki value of 30.1 mM glucose (Cota et al., 2015). Ki value of TnBglB is

greater than many formerly published GH3 fungal β-glucosidases. For instance, β-

glucosidases from Myceliophthora thermophila with Ki of 0.282 mM (Karnaouri et al.,

2013), and Neosartorya fischeri with Ki of 13.4 mM (Yang et al., 2014).

Effect of xylose on activity of TnBglA and TnBglB was scrutinized. TnBglA revealed not

only great tolerance against xylose inhibition although its relative activity was highly

enhanced by 170% with 100 mM xylose. The stimulatory effect of xylose on TnBglA

activity was observed over a broad range of 50-700 mM xylose. Whereas, TnBglB

exhibited less tolerant behavior to xylose and its relative activity was gradually decreased

up to 18% in the presence of 600 mM xylose. TnBglA and TnBglB have a Ki value of 1300

mM and 200 mM xylose, respectively (Figure 4.48b). Despite the fact that minor β-

glucosidases from other bacteria displayed sufferance to xylose. Ki of TnBglA and TnBglB

for xylose inhibition are as high as compared to other xylose-activated β-glucosidase from

Scytalidium thermophilum that had a Ki of 43.24 mM (Zanoelo et al., 2004).

Substrate specificity classifies β-glucosidases into three main groups, the first group is

known as aryl- β-glucosidases which have high affinity to aryl-β-glucosides. Second group

contains true cellobiases which preferably hydrolyze only oligosaccharides to release

glucose, while third group consists of broad specific β-glucosidases that hydrolyze many

types of substrates, and these are the most frequently observed form of β-glucosidases (Pei

et al., 2012). In this present study, affinity of TnBglA and TnBglB was determined over a

large number of heteroglycans, xylan, various para- and ortho-nitrophenyl glycosides

substrates with both α and β confirmation. Both enzymes displayed maximum specificity

towards pNP-β-D-glycosides and cellobiose substrates, while no noticeable activity

Page 251: A THESIS TITLED - prr.hec.gov.pk

Discussion

220

observed with pNP-α-D-glycosides and oNP-glycosides, these results suggested that both

enzymes specific for β-linkage and belong to first group aryl-β-glucosidases. The

hydrolyzing activity for substrates having β-1-4 linkages such as pNPG, pNPF and pNPGal

were much higher than for the substrates involving β-1-2 linkages such as oNPG, oNPF

and oNPGal. The presence of nitro group at the ortho position reduced the catalytic

efficiency, which indicated the steric hindrance with a hydroxyl group at C-6 (Parry et al.,

2001). Both enzymes exhibited no catalytic affinity with sucrose, xylan, carboxymethyl

cellulose (CMC), avicel and laminarin substrates (Table 4.8).

Several studies have been reported that β-glucosidases possess high affinity to aryl-β-

glycosides (pNPG, pNPGal, and pNPX) or β-linkage and the low affinity towards α-

linkage, which is a common property of GH1 (Pei et al., 2012; Fang et al., 2014). However,

a β-glucosidase from Sphinogopyxis alakensis had maximum activity with β-1,2 linked

substrate oNP-β-glucopyranoside (Shin and Oh, 2014). TnBglB has high affinity towards

aryl-β-glycosides of glucose and xylose. Xylosidase activity of TnBglB could contribute to

the degradation of xylan in pretreated plant biomass, and a β-glucosidase having β-

xylosidase activity can enhance the hydrolysis capacity. This phenomenon is consistent

with GH3 β-glucosidases from T. maritium (Goyal et al., 2001), while from Terrabacter

ginsenosidimutans revealed maximum activity towards oNPG instead of pNPG (An et al.,

2010).

Kinetics parameters of both enzymes, Km, Vmax, kcat, and kcat Km-1 for the hydrolysis of

substrates like pNPG, pNPF, pNPGal, pNPX, pNPC and cellobiose were calculated using

Lineweaver–Burk plot (Figure 4.49a and 4.49b), all values are provided in table 4.9a and

4.9b. Kinetic values of both enzymes clarified that pNPG is a more specific substrate than

cellobiose and other substrates. TnBglA has lower Km value (1.5 mM) against pNPG than

GH1 β-glucosidases from Caldicellulosiruptor bescii with Km of 3.71 mM (Bai et al.,

2013), demonstrating higher affinity for pNPG. Conversely, TnBglA exhibited Km of 7.76

mM for cellobiose, where this finding is lower than from Thermoanaerobacterium

aotearoense (Km value of 25.45 mM) (Yang et al., 2015). Catalytic efficiency (kcat Km-1)

ratio is often supposed as to measure enzyme efficiency, large value of kcat and high affinity

Page 252: A THESIS TITLED - prr.hec.gov.pk

Discussion

221

for substrate make kcat Km-1 value large. TnBglA values of Vmax, kcat and kcat Km

-1 for pNPG

and cellobiose are highest from many previous studies (Bai et al., 2013; Yang et al., 2015).

TnBglB revealed the lowest Km value (0.45 mM) for pNPG representing greater affinity for

substrate as compared to the Km values of other bacterial GH3 thermophilic β-glucosidases

included Thermus sp. IB-21 (Kang et al., 2005) Terrabacter ginsenosidimutans (An et al.,

2010), and T. thermarum (Long et al., 2016) that exhibited Km values of 11.8 mM, 4.2 mM

and 2.41 mM, respectively. TnBglB presented the high Km value of 7.35 mM for cellobiose,

which indicated less affinity to the substrate. This finding is in contrast to GH3 β-

glucosidases with low Km values for cellobiose obtained from Phanerochaete

chrysosporium (Km of 5.05 mM) (Kawai et al., 2003), and Penicillium funiculosum NCL1

(Km of 1.25 mM) (Ramani et al., 2015) had been reported earlier. TnBglB has the highest

kcat and kcat Km-1 values for pNPG as compared to the values for GH3 β-glucosidases from

T. neapolitana (Pozzo et al., 2010), Dictyoglomus turgidum (Kim et al., 2011), and T.

petrophila (Cota et al., 2015).

Genomic analysis of various thermotolerant β-glucosidases revealed that amino acid

composition is the fundamental factor which associated with the thermostability of protein,

when compared to their heat labile or mesophilic counterparts. Usually heat loving β-

glucosidase have more hydrophobic and helix forming residues, and less thermolabile

residues (Suhre and Claverie, 2003; Gupta et al., 2014). In general, the stability of β-

glucosidase increases with the insertion of helix-forming residues into α-helices, and

stability decreases with the addition of helix-breaking residues (Singh and Hayashi, 1995).

There are 193 helix-forming residues present out of 446 amino acids in TnBglA, and 233

helix-forming residues present out of 721 residues in TnBglB, suggesting this could be one

of the factor influencing the thermal stability.

Thermostability of β-glucosidase is enhanced in the presence of high percentage of charged

glutamic acid (E) and proline (P) residues (Matthews et al., 1987) whereas, lower

percentage of glutamine (Q) along with neutral residues methionine (M) and cysteine (C)

account for the formation of salt-bridges/ion pairs and H-bonds which play important role

in structure stabilization. Earlier, Voorhorst and colleagues (1995) described that an

Page 253: A THESIS TITLED - prr.hec.gov.pk

Discussion

222

increase in acidic amino acids residues particularly glutamic acid (E) and a decrease in

number of neutral residues notably cysteine (C), are thought to the account for heat stability

of enzyme. Godde et al. (2005) also demonstrated that increases proportion of acidic

residues enhance the optimal temperature of enzyme. Thermotolerant TnBglA and TnBglB

have high percentage of charged residues, 63 and 116 negatively charged residues (aspartic

acid and glutamic acid) while 51 and 94 positively charged residues (arginine and lysine),

respectively.

TnBglA protein contains only 1 cysteine (C), 29 aspartic acid (D) and 34 glutamic acid (E)

residues, however TnBglB protein has only 4 cysteine (C), 41 aspartic acid (D) and 75

glutamic acid (E) residues, which is fairly similar to other thermophilic β-glucosidases

(Jabbour et al., 2012). Both proteins have lower frequency of hydrophilic and thermolabile

residues glutamine (Q) due to its propensity to undergo deamination, methionine (M) and

cysteine (C) due to their tendency to undergo oxidation at high temperature (Gupta et al.,

2014). Overall high hydrophobicity is also influenced the stability (Voorhorst et al., 1995).

TnBglA and TnBglB have 66 and 69 aromatic residues, about 15% and 10% of the

sequences, respectively. On average, the sequence of non-thermostable β-glucosidases

randomly picked from NCBI database, are composed of 6% aromatic acids. This finding

is in accord to the analysis of thermostable β-glucosidase from Fervidobacterium

islandicum (Jabbour et al., 2012).

Further non-polar residues such as alanine (A) regard as one of the best helix forming

residue, presence of isoleucine (I) and leucine (L) and valine (V) provide the

conformational stability and also contribute to enhance the thermostability of globular

proteins. Thermostable TnBglA and TnBglB have higher values of aliphatic index 82.60

and 84.17, respectively which is due to high percentage of non-polar residues (aliphatic

side chains), is a statistical approach used to determine enzyme thermostability by Ikai

(1980). Aliphatic index is defined as the relative volume occupied by aliphatic side chains

(alanine, leucine, valine, and isoleucine) which is conspicuously higher for thermophilic as

compared to mesophilic proteins.

Page 254: A THESIS TITLED - prr.hec.gov.pk

Discussion

223

Stability can also be estimated by instability index, which plays an essential role in

determining half-life of a protein. A stable protein has an instability index less than 40,

while a protein is predicted to be unstable if its value is above 40 (Gupta et al., 2014).

TnBglA and TnBglB have 31.23 and 30.83 instability index, respectively. Mostly

thermostable β-glucosidases have instability index below 40 which makes them heat-stable

according to the study of Guruprasad et al. (1990). Grand average hydropathicity

(GRAVY) measures the hydropathicity of a specific protein, lower the GRAVY value,

better will be the protein interaction with water. TnBglA and TnBglB have -0.373 and -

0.427 values of GRAVY because all thermophilic proteins have low GRAVY value (Gupta

et al., 2014).

Aliphatic index, instability index and GRAVY values of TnBglA and TnBglB are in

accordance to the several heat tolerant GH1 and GH3 β-glucosidases and comparable to

the thermolabile β-glucosidases. For instance, GH1 thermophilic β-glucosidase T.

neapolitana had 80.99 aliphatic index, 33.45 instability index and -0.423 GRAVY values

(Park et al., 2005). Whereas, a mesophilic GH1 β-glucosidase from Bifidobacterium breve

displayed 77.63 aliphatic index, 26.75 instability index and -0.330 GRAVY (Nunoura et

al., 1996). Thermophilic GH3 β-glucosidase from Clostridium thermocellum had 89.59

aliphatic index, 37.14 instability index and -0.310 GRAVY values (Graebnitz et al., 1989),

which are comparable with TnBglB. Whereas, thermolabile GH3 β-glucosidase from

Terrabacter ginsenosidimutans showed 88.92 aliphatic index, 42.37 instability index and

-0.198 GRAVY (An et al., 2010).

Hyperthermophilic proteins (β-glucosidases) have more intramolecular interactions (H-

bonding, hydrophobic interactions, electrostatic interactions and disulfide bonding), strong

network of salt bridges, higher packing efficiency, greater rigidity, increased helical

content, shortened/less loops, conformation strain release and reduced entropy of unfolding

than mesophilic proteins which increase their stability (Li et al., 2005; Gupta et al., 2014).

In this study, the inherent thermostability of TnBglA and TnBglB is further verified and

corroborated by their kinetic and thermodynamic study.

Page 255: A THESIS TITLED - prr.hec.gov.pk

Discussion

224

Thermodynamic study of TnBglA and TnBglB was performed to measure the

thermostability of substrate hydrolysis system. Transition state stabilization was described

using an alternative approach as a description for the rate enhancement with assay

temperature (Benkovic and Hammes-Schiffer, 2003). Thermodynamic parameters: Gibbs

free energy (ΔG*), enthalpy (ΔH*), entropy (ΔS*) and activation energy (Ea) for pNPG

hydrolysis by TnBglA and TnBglB were calculated using Arrhenius approach in a

temperature range from 80°C to 95°C and 70°C to 85°C, respectively (Table 4.11a and

4.11b). The result displayed that both enzymes have a highly thermotolerant system

because these parameters are nearly constant at various temperatures. Negative entropy and

low enthalpy values explain the formation of ordered and effective enzyme substrate

transition state complex (Akolkar and Desai, 2010). These parameters at high temperature

are comparable to the thermodynamic studies carried out for thermostable β-glucosidases

from Fusarium solani (Bhatti et al., 2013) and Thermotoga maritima (Mehmood et al.,

2014).

Thermal stability of protein (β-glucosidases) is relevant for biological function and

molecular evolution. Although complex, the thermal denaturation process of TnBglA and

TnBglB can be simplified as the classical two-step process in which the first step involves

unfolding of the protein's native structure. The unfolded polypeptide may refold reversibly

to the native conformation or in a second step undergo irreversible denaturation to

permanent inactivation due to protein aggregation, misfolding and covalent changes such

as the deamidation of asparagine (N) or glutamine (Q) residues and oxidation of cysteine

(C) or methionine (M) residues (Volkin and Klibanov, 1989). This denaturation is

accompanied by the disruption of non-covalent linkages, including hydrophobic

interactions, with concomitant increase in the enthalpy of activation (ΔH*D) and increase

in the disorder or entropy (ΔS*D) of activation (Marín et al., 2003).

Enzyme thermostability encompasses thermodynamic and kinetic stabilities (Sanchez-

Ruiz, 2010; Bommarius and Paye, 2013). Thermodynamic stability is governed by the

enzyme free-energy of stabilization, reflecting the difference between the free energies of

the folded and the unfolded states of the protein, and by its melting temperature (Tm,

temperature at which 50% of the protein is unfolded). Kinetic or long-term stability

Page 256: A THESIS TITLED - prr.hec.gov.pk

Discussion

225

depends on the energy barrier to irreversible inactivation and is generally expressed as the

enzyme's half-life (t1/2) at a defined temperature. An increase in the enzyme resistance to

unfolding (higher Tm) also increases its resistance to inactivation (higher t1/2) as increase in

the stability of the native state leads to slower accumulation of the unfolded state and

secondly, the unfolded state is usually the ground state leading to irreversible denaturation

or inactivation.

In order to determine the thermodynamic parameters for thermal stability, energy of

activation (EaD) for thermal denaturation of TnBglA and TnBglB were determined by

applying the Arrhenius plot. The plot of residual activity versus incubation time in a

temperature range of 97°C to 99°C for TnBglA and 94°C to 96°C for TnBglB are linear

(R2>95%) indicating first order kinetics of inactivation (Figure 4.51a and 4.51b). With

increasing temperature, t1/2 value decreased and the first order deactivation rate constant

(Kd) increased indicating that enzyme is less thermostable at high temperature. EaD values

for TnBglA and TnBglB are found to be very high, 665.12 kJ mol-1 and 286.83 kJ mol-1,

respectively. These values indicating both enzymes are very stable and compact, which are

highly resistant to heat denaturation. The higher value of EaD means that more energy is

required to denature the enzyme (Tayefi-Nasrabadi and Asadpour, 2008) and that the

enzyme might have undergone considerable conformational changes during denaturation

(Marín et al., 2003).

The value of free energy of thermal denaturation (ΔG*D) for TnBglA and TnBglB decreased

with the increase in temperature as shown in table 4.12a and 4.12b, respectively. When

entropy of inactivation (ΔS*D) of TnBglA and TnBglB was calculated at each temperature,

it showed positive values, which indicates that there are no significant processes of

aggregation, since had this happened, the values would have been negative (Marín et al.,

2003). TnBglA and TnBglB established high stability and displayed a half-life (t1/2) of 5.21

and 4.44 minutes at temperature 97°C and 94°C, respectively. Thermodynamic analysis of

TnBglA and TnBglB are in line with some recent studies of recombinant enzymes (endo-

1,4-β-xylanase and endo-1,4-β-glucanase) from T. petrophila (Haq et al., 2012a; Haq et

al., 2015a). However, limited literature is available on the thermodynamic study of

recombinant β-glucosidases.

Page 257: A THESIS TITLED - prr.hec.gov.pk

Discussion

226

The predicted secondary structure of TnBglA has 42% helices (H), 10% β-sheet (E) and

47% loop (C). Three-dimension structure modeling of monomeric TnBglA was elucidated

by GENO-3-D and validated by Procheck. Ramachandran plot suggested that 92.5%

residues are in the most favorable regions (Figure 4.54a). The predicted 3-D structure

showed that it consists of 12 α-helices (major) which is a common motif in hydrolases

secondary structure and has 12 β-sheets, in which 8 β-sheets are present in the core and

making barrel shape structure while 4 β-sheets are at the surface of the protein (Figure

4.55a). The predicted structure of TnBglA represents an interesting pattern of conserved

structural elements in the way that outer part of the structural fold contains α-helices

whereas inner part contains approximately all parallel β-sheets form catalytic cleft or

pocket.

The α-helices and β barrel are connected with each other through a chain that provides

structural stability and active site is placed at top face of the barrel. One of the structural

characteristic is that the loops at C-terminal end of the sheets frequently involve in active

site of protein (Wierenga, 2001). TnBglA has conserved structural loops and classical (α/β)8

triose phosphate isomerase (TIM) barrel structure/scaffold, commonly found in proteins,

span in cell membranes and binds hydrophobic ligands in the barrel center. The TIM barrel

scaffold is one of the key characteristics and highly conserved fold of GH1 protein and

probably the most commonly present folding motif in proteins from mesophiles to

thermophiles (Branden, 1991; Wierenga, 2001). Structural modeling studies of TnBglA

demonstrated that it is quite similar to GH1 β-glucosidases such as from Fervidobacterium

islandicum (Jabbour et al., 2012), and Thermoanaerobacterium aotearoense (Yang et al.,

2015).

In the case of GH1 β-glucosidases, the two essential catalytic glutamic acids (E) are about

200 residues away from each other in the sequence. While, in TnBglA, catalytic acid/base

residue E166 is located in the conserved TLNEP motif at C-terminal end of β-strand 4, and

nucleophile catalysts of TnBglA are E351 and E405, located in the conserved ITENG and

DNFEW motifs at C-terminal end of β-strand 7 (Henrissat et al., 1995; Jenkins et al.,

1995). Active-site glutamic acids (Glu) are present in a pocket shaped structure with

hydrophobic residues at the entrance point (Gasteiger et al., 2003). The distance between

Page 258: A THESIS TITLED - prr.hec.gov.pk

Discussion

227

the active glutamic acid E166 and E351 is 4.7 Aͦ and distance between E166 and E405 is 10.1 Aͦ

(Figure 4.55b). TnBglA is typical of a retaining β-glucosidase and permits for the formation

of a glycosyl-enzyme intermediate (McCarter and Withers, 1994).

The predicted secondary structure of TnBglB is composed of 25% helices (H), 19% β-sheet

(E) and 55% loop (C). Three-dimension profile structure modeling of TnBglB was

elucidated by GENO-3-D and validated by Procheck. Ramachandran plot statistics of

TnBglB suggested that 92.3% residues are in the most favorable regions (Figure 4.54b).

The predicted 3-D structure of TnBglB showed that enzyme consists of three domains

including an (α/β)8 TIM barrel domain-I, 5-stranded α/β sandwich domain-II, and

fibronectin type III (FnIII) domain is sequentially inserted between first two domains

(Figure 4.55c). At C-terminal, 120 residues are involved to form a FnIII domain and

believed to provide heat stability to GH3 β-glucosidases by managing the relative

movement of domain I and II, though all three domains are connected to each other by two

linkers regions that provides structural stability and play key role in folding of protein into

its active form (Ramírez-Escudero et al., 2016).

The structural fold and catalytic cleft for substrate of TnBglB is highly conserved with

other GH3 β-glucosidases in the evolutionary history. All part of TnbglB structure are well

organized and allows all residues involved in hydrolytic activity to be identified. The

active-site residues are found at the interface of domain-I and II, and formed a catalytic

pocket or crater which is very large and good enough to accommodate large substrates.

Domain-I (α/β)8 TIM barrel fold is highly conserved and probably the most frequently

present folding motif in GH3 proteins from mesophiles to thermophiles. Surprisingly, all

β-strands in domain-I are not parallel as would be predicted in a TIM barrel, the direction

of some β-strands is reversed (anti parallel) with respect to TIM barrel topology in general

(Pozzo et al., 2010). FnIII domain is located on TnBglB side remotest from the active

region, its actual function has rarely been demonstrated unambiguously. Domain-III have

been found previously in a number of bacterial extracellular GHs displaying different

specificities including chitinases, pullulanases and cellobiohydrolases (Irwin et al., 2000;

Kataeva et al., 2002).

Page 259: A THESIS TITLED - prr.hec.gov.pk

Discussion

228

In 3-D structure of TnBglB, domain-I contains a catalytic nucleophile aspartic acid (D242),

which is located in the conserved GFVMSDW region at C-terminal. However, a catalytic

acid/base glutamic acid residue (E458) is located in a conserved SGEG region of domain-

II, both active-site residues aspartic acid (D242) and glutamic acids (E458) are present in an

organized pocket shaped structure with hydrophobic amino acid residues at the entrance

point leading to the catalytic pocket (Pozzo et al., 2010). The distance between nucleophile

D242 and acid/base residue E458 is 5.8 Aͦ (Figure 4.55d). TnBglB like other GH3 β-

glucosidases has a retaining catalytic mechanism, the main catalytic function of enzyme is

to hydrolyze β-linkages between glucose and other moieties of diverse substrates structure

(Turner et al., 2006; Turner et al., 2007). From the structural modeling studies, it is

concluded that TnBglB has conserved structural loops and identical folds, highly analogous

to other GH3 β-glucosidases as from T. neapolitana (Pozzo et al., 2010), T. petrophila (Xie

et al., 2015).

Docking studies of TnBglA by PatchDock, more than 100 possible solution of docking

were predicted, which were evaluated through Procheck and further analysed the enzyme-

substrate complexes by ligplot. All of the best docking models showed catalytic mechanism

of TnBglA in which at least two or three conserved glutamic acid residues (Glu166, Glu351

and Glu405) were found in close contact to the substrate for hydrolysis. Docking data of

enzyme-substrate also in harmony with the earlier findings of GH1 β-glucosidases (Gloster

et al., 2007; Haq et al., 2012b). Furthermore, with cellobiose W324 (Tryptophan), F414

(Phenylalanine), W398 (Tryptophan), W122 (Tryptophan), W406 (Tryptophan), H180

(Histidine), E408 (Glutamic acid) and M322 (Methionine) amino acid residues present on the

periphery of the reaction complex (Figure 4.56a). Whereas, in case of pNPG, E166

(Glutamic acid), W398 (Tryptophan), W324 (Tryptophan), W122 (Tryptophan), W406

(Tryptophan), H180 (Histidine) and E408 (Glutamic acid) residues present on the periphery

of reaction complex (Figure 4.56b). These residues provide a hydrophobic surrounding to

improve reactivity and thermostability, as suggested previously for GH1 thermostable β-

glucosidases (Bai et al., 2013; Rajoka et al., 2015; Yang et al., 2015).

Docking studies of TnBglB by PatchDock, more than 100 possible solution of docking

were generated, which were evaluated through Procheck and additionally analysed the

Page 260: A THESIS TITLED - prr.hec.gov.pk

Discussion

229

enzyme-substrate complexes by ligplot. All of the best docking models showed that

cellobiose and pNPG substrates can be docked into the catalytic pocket of TnBglB very

well, in which D58 (Aspartic acid), R64 (Arginine), E458 (Glutamic acid) and D242 (Aspartic

acid) active-site residues are involved in catalytic mechanism of TnBglB and tightly locked

the substrates in a network of hydrogen bonds (Figure 4.57a and 4.57b). However, two

highly conserved essential catalytic residues are found in close contact to the both

substrates, in which D242 (Aspartic acid) act as nucleophile and E458 Glutamic acid as an

acid/base in the hydrolytic reactions. Furthermore, side chain of S370 (Serine), L25

(Leucine), L28 (Leucine) and W243 (Tryptophan) residues present on the periphery of

reaction complex and may form weak hydrogen bond with cellobiose. Likewise, the

surrounding residues M207 (Methionine), S370 (Serine), L25 (Leucine), L116 (Leucine) and

W243 (Tryptophan) may form weak hydrogen bond with pNPG. These residues provide a

hydrophobic platform to enhance thermal stability and reactivity. This finding is largely

congruent with the docking results of GH3 β-glucosidases from T. neapolitana (Pozzo et

al., 2010) and ruminal microbe (Ramírez-Escudero et al., 2016).

Enhancing the production, expression and activity of heterologous proteins in microbial

host expression system is the most prominent aspect of modernized and revolutionized

biochemistry (Rosano and Ceccarelli, 2014). Therefore, production and expression of

TnBglA and TnBglB were enhanced by using modifying media and lactose inducer instead

of IPTG. All cultivation parameters were optimized again with all inducing media as

described above such as pre-induction optical cell density, growth temperature, medium

composition, better agitation, heat shock treatment (at 42°C) to the recombinant culture

and inducer concentration.

Hyperthermophilic β-glucosidases from T. naphthophila (TnBglA and TnBglB) are highly

effective and valuable proteins, hence to augment the volumetric yield of desired products

and bacterial cultures, ten various nutrient complex and modified inducing media (LB,

LB+, ZB, ZBM, ZYB, 3×ZYB, 4×ZB, M9, ZYBM9, and 3×ZYBM9) were used. Induced

them individually with different concentration of IPTG and lactose at appropriate induction

point. Mostly, all these media were designed for T7 expression system using pET vectors

in E. coli BL21 (Studier, 2005). Individually, comparative analysis of IPTG and lactose

Page 261: A THESIS TITLED - prr.hec.gov.pk

Discussion

230

inducers in above mentioned media revealed that the maximum culture production and

activity of both enzymes were obtained in 4×ZB medium with 0.5 mM IPTG and 150 mM

lactose after 72 h induction at 22°C with 200 rev min-1 (Figure 4.58 and 4.59).

The extracellular activity of TnBglA is enhanced in 4×ZB medium by 3.74 fold with 0.5

mM IPTG and 3.8 fold with 150 mM lactose as compared to the LB medium induced with

IPTG in a shake flask. Whereas, extracellular TnBglB activity is enhanced by 0.096 and

0.085 fold in 4×ZB medium using lactose and IPTG inducers, respectively. Approximately

same level of TnBglB expression was observed with both inducers. Similarly, the

production and expression in term of activity of several recombinant proteins have been

enhanced by applying optimal cultivation parameters and lactose induction strategy (Cheng

et al., 2011; Hameed et al., 2014; Su et al., 2015). The inducing modified medium, 4×ZB

is a tryptone rich nutritional medium, which help host cells growth and improve the

heterologous protein production. Similarly, Basar et al. (2010) has been reported that

thermophilic cloned enzyme activity and E. coli culture production was enhanced by using

complex cultivation media.

However, IPTG is significantly effective and a stable inducer, commonly used in

laboratory to induce T7 lac promoter for the heterologous proteins expression in host but

must be removed from the targeted proteins due to its potential toxicity. Lactose inducer

of lac operon is quite different from IPTG, it causes less metabolic burden on host (E. coli

BL21) machinery, acts as carbon and energy source to promote growth and increase the

culture density, and may improve the output of desired protein. Lactose, unable to enter

the host cells, requires an enzyme primase which converts into allolactose by β-

galactosidase and finally able to start T7 lac promoter. Lactose inducing procedure is

complex than IPTG, therefore if lactose is selected as inducer, then inducing parameters

(inducer concentration and incubation time, etc.) must be optimized. It has been proved

that IPTG is highly toxic for bacterial growth whereas lactose is an alternative natural,

efficient, non-toxic and inexpensive inducer which has numerous incredible advantages

and successfully use for the industrial production of engineering products (Kilikian et al.,

2000; Yan et al., 2004).

Page 262: A THESIS TITLED - prr.hec.gov.pk

Discussion

231

In the present study, after lactose induction, the growth of cloned TnBglA expressing

bacteria was high than that achieved with IPTG, however maximum culture density of

18.96 with 11.30 g DCW L-1 was acquired in 4×ZB medium induced with 150 mM lactose

(Table 4.14b). In all inducing media, growth of recombinant TnBglB producing bacteria

was high with lactose induction as compared to IPTG, and maximum culture density of

18.66 with 11.08 g DCW L-1 was achieved in 4×ZB medium with 150 mM lactose

induction (Table 4.14b). This behavior is according to the previous study of enhanced

production of cloned enzyme from Pyrococcus furiosus (Ikram et al., 2009) and Bacillus

halodurans (Naz et al., 2010).

The yield of TnBglA and TnBglB greatly improved when cultures were given a heat shock

at 42°C (1 h) just before IPTG/lactose induction followed by incubation at 22°C for 72 h

in a shaking incubator (200 rev min-1), in all inducing media the similar results were

observed as in case of LB medium induced with IPTG (described earlier in this chapter).

Induction strategy benefits the balance of bacterial growth, as well as cloned protein

synthesis, folding and transportation. Optimal TnBglA and TnBglB expression observed

with 0.5 mM IPTG in all media including optimal 4×ZB; and 150 mM lactose determined

to be ideal concentration for induction in all media including 4×ZB. Great amount of

allolactose is needed at the time of induction, thus more amount of lactose in growth

medium is mandatory. Similar strategy of lactose induction has been applied to enhance

the expression of proteins in engineered E. coli BL21 (Tran et al., 2010; Bashir et al., 2015;

Su et al., 2015).

Highly thermostable, xylose and glucose-tolerant cloned TnBglA and TnBglB expectedly

increases the competency of monosugar (glucose) production by synergistic interaction

with other thermostable cellulases (EGs and CBHs) during saccharification process of

biomass (data not shown) for consequent production of bioethanol. Saccharification of

pretreated biomass (bagasse, wheat bran and rice straw) is highly improved, when either

TnBglA or TnBglB used with other enzymes in cocktail, indicating both are strong

candidates for the bioconversion process. Use of these enzymes, with a low degree of

inhibition might improve the yield and reduce the cost of process, and also have several

biotechnological and industrial applications.

Page 263: A THESIS TITLED - prr.hec.gov.pk

CHAPTER-VI

CONCLUSION

Page 264: A THESIS TITLED - prr.hec.gov.pk

Conclusion

232

6. Conclusion

As biocatalysts and analytical tools, thermozymes have to date gained a great deal of

interest as novel enzymes with unique properties, and are expected to fill the gap between

biological and chemical industrial processes. In this present research work, focus was on

the development of such industrially relevant thermostable and persuasive enzymes.

Therefore, two novel cellulolytic genes (TnbglA and TnbglB) from a hyperthermophilic

bacterium T. naphthophila RKU-10T, were cloned and overexpressed in E. coli BL21

CodonPlus (DE3)-RIPL, a mesophilic expression host. Sequencing and alignment studies

demonstrated that β-glucosidase A (TnBglA) and β-glucosidase B (TnBglB) are belonged

to GH family 1 and 3, respectively. Several cultivation and induction strategies were

adopted to achieve the optimal production and expression of recombinant TnBglA and

TnBglB. Culturing in optimal modified medium (4×ZB) and induction with lactose (150

mM) significantly enhanced the transformant growth (optical density) and heterologous

expression of both proteins. Hence lactose in contrast to IPTG, has proved as an efficient,

cheap and non-toxic alternative inducer. It has also recognized that optimal fermentation

conditions such as pre-induction optimal density, heat shock treatment, inducer

concentration, agitation, pH of medium, inducement temperature and time, would be

valuable aspect to increase the production, expression and proper folding of recombinant

proteins. This research provides two useful novel β-glucosidases (TnBglA and TnBglB)

which displayed favorable properties as high thermostability, glucose and xylose tolerance,

independence of metallic cations, and high catalytic activity. All these prominent features

make both enzymes potential candidates to be used in SSF processes for production of

bioethanol as well as suitable for paper, textile, pharmaceutical, cosmetic, animal feed,

food and juices, and these enzymes have many other industrial and biotechnological

applications.

Page 265: A THESIS TITLED - prr.hec.gov.pk

REFERENCE

Page 266: A THESIS TITLED - prr.hec.gov.pk

References

233

References

Akolkar A.V., and A.J. Desai. 2010. Catalytic and thermodynamic characterization of

protease from Halobacterium sp. SP1 (1). Res. Microbiol., 161: 355-362.

An, D-S., C.H. Cui, H.G. Lee, L Wang, S.C. Kim, S.T. Lee, F. Jin, H. Yu, Y.W. Chin, H.K.

Lee, W.T. Im, and S.G. Kim. 2010. Identification and Characterization of a Novel

Terrabacter ginsenosidimutans sp. nov. β-glucosidase that transforms Ginsenoside

Rb1 into the rare Gypenosides XVII and LXXV. Applied and Environmental

Microbiology, 76(17): 5827–5836.

Artzi, L., B. Dassa, I. Borovok, M. Shamshoum, R. Lamed, E.A. Bayer. 2014.

Cellulosomics of the cellulolytic thermophile Clostridium clariflavum. Biotechnol

Biofuels. 7:100.

Bai, A., X. Zhao, Y. Jin, G. Yang, and Y. Feng. 2013. A novel thermophilic β-glucosidase

from Caldicellulosiruptor bescii: Characterization and its synergistic catalysis with

other cellulases. J. of Molecular Catalysis B: Enzymatic. 85: 248–256.

Bajaj, B.K., H. Pangotra, M.A. Wani, A. Sharma, and P. Sharma. 2009. Characterization

of thermo-tolerant and acid/alkali tolerant β-glucosidase from bacterial isolate M+.

Journal of scientific and industrial research, 68(3): 242-247.

Bankova, E., N. Bakalova, S. Petrova, and D. Kolev. 2006. Enzymatic synthesis of

oligosaccharides and alkylglycosides in waterorganic media via transglycosylation

of lactose. Biotechnol. Biotechnol. Equip., 20(3): 114–119.

Barnard, D., A. Casanueva, M. Tuffin, and D. Cowan. 2010. Extremophiles in biofuel

synthesis, Environ. Technol., 31(8-9): 871–888.

Basar, B., M.M. Shamzi, M. Rosfarizan, N.N.T. Puspaningsih, and A.B. Ariff. 2010.

Enhanced production of thermophilic xylanase by recombinant Escherichia coli

DH5α through optimization of medium and dissolved oxygen level. Int. J. Agri.

Biol., 12(3): 321-328.

Bashir, H., N. Ahmed, A.U. Zafar, M.A. Khan, S. Tahir, M.I. Khan, F. Khan, and T.

Husnain. 2015. Simple procedure applying lactose induction and one step

purification for high yield production of rhcifn. Biotechnol. Appl. Biochem., doi:

10.1002/bab.1426.

Page 267: A THESIS TITLED - prr.hec.gov.pk

References

234

Bazmi, A.A., G. Zahedi. 2011. Sustainable energy systems: Role of optimization modeling

techniques in power generation and supply–A review. Renew Sustain Energy Rev.,

15(8): 3480–3500.

Belancic, A., Z. Gunata, M.J. Vallier, and E. Agosin. 2003. β-glucosidase from the grape

native yeast Debaryomyces vanriji: Purification, characterization and its effect on

monoterpene concentration of a Muscat grape juice. J. Agri. Food Chem., 51(5):

1453-1459.

Benkovic, S.J., and S. Hammes-Schiffer. 2003. A perspective on enzyme catalysis.

Science, 301: 1196–1202.

Bhalla, A., N. Bansal, S. Kumar, K.M. Bischoff, and R.K. Sani. 2013. Improved

lignocellulose conversion to biofuels with thermophilic bacteria and thermostable

enzymes. Bioresour. Technol., 128: 751–759.

Bhatia, Y., S. Mishra, and V.S. Bisaria. 2002. Microbial beta-glucoidases: cloning,

properties and applications. Crit. Rev. Biotechnol., 22(4): 375–407.

Bhatti, H.N., S. Batool, and N. Afzal. 2013. Production and characterization of a novel β-

glucosidase from Fusarium solani. Int J Agric Biol., 15: 140‒144.

Blumer-Schuette, S.E., I. Kataeva, J. Westpheling, M.W. Adams, and R.M. Kelly. 2008.

Extremely thermophilic microorganisms for biomass conversion: status and

prospects. Curr. Opin. Biotechnol., 19: 210–217.

Bommarius, A.S., and M.F. Paye. 2013. Stabilizing biocatalysts. Chem. Soc. Rev. 42(15):

6534–6565.

Bradford, M.M. 1976. A rapid and sensitive method for the quantitation of microgram

quantities of protein utilizing the principle of protein-dye binding. Anal Biochem.,

72: 248-54.

Branden, C.-I. 1991. The TIM barrel–the most frequently occurring folding motif in

proteins. Curr. Opin. Stuct. Biol., 1(6): 978–983.

Breves, R., K. Bronnenmeier, N. Wild, F. Lottspeich, W.L. Staudenbauer, and J.

Hofemeister. 1997. Genes encoding two different beta-glucosidases of

Thermoanaerobacter brockii are clustered in a common operon. Appl. Environ.

Microbiol., 63(10): 3902-10.

Page 268: A THESIS TITLED - prr.hec.gov.pk

References

235

Bringezu, S., H. Schütz, M. O´Brien, L. Kauppi, R.W. Howarth, J. McNeely. 2009.

Towards sustainable production and use of resources: Assessing biofuels. UNEP;

http://www.unep.org/PDF/Assessing_Biofuels.pdf.

Bronnenmeier, K., and W. Staudenbauer. 1988. Purification and properties of an

extracellular beta-glucosidase from the cellulolytic thermophile Clostridium

stercorarium. Appl. Microbiol. Biotechnol., 28(4): 380-386.

Bu, L., M.F. Crowley, M.E. Himmel, and G.T. Beckham. 2013. Computational

investigation of the pH dependence of loop flexibility and catalytic function in

glycoside hydrolases. J. Biol. Chem., 288(17): 12175-86.

Cairns, J.R.K., and A. Esen. 2010. β-Glucosidase. Cell Mol. Life Sci., 67(20): 3389-3405.

Cantarel, B.L., P.M. Coutinho, C. Rancurel, T. Bernard, V. Lombard, and B. Henrissat.

2009. The Carbohydrate-Active EnZymes database (CAZy): an expert resource for

glycogenomics. Nucleic Acids Res., 37(1): 233-238.

Chan, C.S., L.L. Sin, K.G. Chan, M.S. Shamsir, F.A. Manan, R.K. Sani and K.M. Goh.

2016. Characterization of a glucose-tolerant β-glucosidase from Anoxybacillus sp.

DT3-1. Biotechnology for Biofuels, 22(9): 174.

Chang, J., I-H. Park, Y-S. Lee, S-C Ahn, Y. Zhou, and Y-L Choi. 2011. Cloning,

expression, and characterization of β-glucosidase from Exiguobacterium sp. DAU5

and transglycosylation activity. Biotechnol. Bioprocess Eng., 16(1): 97–106.

Chen, R., Y.Z. Wang, Q. Liao, X. Zhu, T.F. Xu. 2013. Hydrolysates of lignocellulosic

materials for biohydrogen production, BMB Rep., 46: 244–251.

Cheng, J., D. Wu, S. Chen, J. Chen, and J. Wu. 2011. High-level extracellular production

of alpha-cyclodextrin glycosyl transferase with recombinant Escherichia coli BL21

(DE3). J. Agric. Food. Chem., 59: 3797-3802.

Chuankhayan, P., T. Rimlumduan, J. Svasti, and J.R. Cairns. 2007. Hydrolysis of soybean

isoflavonoid glycosides by Dalbergia β-glucosidases. J. Agric. Food Chem., 55(6):

2407-2412.

Cohen, S.N., A.C.Y. Chang, and L. Hsu. 1972. Non-chromosomal antibiotic resistance in

bacteria: Genetic transformation of Escherichia coli by R-factor DNA. Proc. Natl.

Acad. Sci., 69: 2110-2114.

Page 269: A THESIS TITLED - prr.hec.gov.pk

References

236

Colussi, F., V.M. Da Silva, I. Miller, J. Cota, L.C. De Oliveira, M.N. De Oliveira, F.M.

Squina, and W. Garcia. 2015. Oligomeric state and structural stability of two

hyperthermophilic β-glucosidases from Thermotoga petrophila. Amino Acids,

47(5): 937-48.

Colussi, F., W. Garcia, E.R. Rosseto, B.L. de Mello, N.M. de Oliveira, I. Polikarpov. 2012.

Effect of pH and temperature on the global compactness, structure, and activity of

cellobiohydrolase Cel7A from Trichoderma harzianum. Eur Biophys J., 41(1):89-

98.

Cota, J., T.L.R. Corrêa, A.R.L. Damásio, J.A. Diogo, Z.B. Hoffmam, W. Garcia, L.C.

Oliveira, R.A. Prade, and F.M. Squina. 2015. Comparative analysis of three

hyperthermophilic GH1 and GH3 family members with industrial potential. New

Biotechnol., 32(1): 13-20.

Dan, S., I. Marton, M. Dekel, B.A. Bravdo, S. He, S.G. Withers, O. Shoseyov. 2000.

Cloning, expression, characterization, and nucleophile identification of family 3,

Aspergillus niger beta-glucosidase. J. Biol. Chem. 275(7): 4973-80.

Daniel, R.M., M.E. Peterson, M.J. Danson, N.C. Price, S.M. Kelly, C.R. Monk, C.S.

Weinberg, M.L. Oudshoorn, and C.K. Lee. 2010. The molecular basis of the effect

of temperature on enzyme activity. Biochem. J., 425(2): 353-360.

Dantur, K.I., R. Enrique, B. Welin, and A.P. Castagnaro. 2015. Isolation of cellulolytic

bacteria from the intestine of Diatraea saccharalis larvae and evaluation of their

capacity to degrade sugarcane biomass. AMB Express, 5: 15.

Decker, C.H., J. Visser, and P. Schreier. 2001. β-glucosidase multiplicity from Aspergillus

tubingensis CBS 643.92: purification and characterization of four β-glucosidases

and their differentiation with respect to substrate specificity, glucose inhibition and

acid tolerance. Appl. Microbiol. Biotechnol., 55(2): 157–163.

Demain, A.L., M. Newcomb, and J.H. Wu. 2005. Cellulase, Clostridia, and Ethanol.

Microbiol. Mole. Biol. Rev., 69(1): 124-154.

Demirbas, A. 2007. Progress and recent trends in biofuels. Progr. Energy Combust. Sci.,

33(1): 1-18.

Page 270: A THESIS TITLED - prr.hec.gov.pk

References

237

De-Moraes, L.M.P., S.A. Filho, and C.J. Ulhaa. 1999. Purification and some properties of

an alpha- amylase and gluco-amylase fusion protein from Saccharomyces

cerevisae. World J. Micro. Biot., 15: 561–564.

Dikshit, R., and P. Tallapragada. 2015. Partial Purification and Characterization of β-

glucosidase from Monascus sanguineus. Braz. Arch. Biol. Technol., 58(2): 185-

191.

Dong, J., Y. Hong, Z. Shao, and Z. Liu. 2010. Molecular cloning, purification, and

characterization of a novel, acidic, pH-stable endoglucanase from Martelella

mediterranea. Journal of microbiology., 48(3): 393–398.

Downing, M., L.M. Eaton, R.L. Graham, M.H. Langholtz, R.D. Perlack, A.F. Turhollow,

B. Stokes, and C.C. Brandt. 2011. US Billion-Ton Update: The Biomass Supply for

a Bioenergy and Bioproducts Industry. Oak Ridge, Tennessee: Oak Ridge National

Laboratory.

Elleuche, S., C. Schafers, S. Blank, C. Schroder, and G. Antranikian. 2015. Exploration of

extremophiles for high temperature biotechnological processes. Curr. Opin.

Microbiol., 25: 113–119.

Elleuche, S., C. Schröder, K. Sahm, G. Antranikian. 2014. Extremozymes-biocatalysts

with unique properties from extremophilic microorganisms. Curr. Opin.

Biotechnol., 29:116-23.

Endler, A., S. Persson. 2011. Cellulose Synthases and Synthesis in Arabidopsis. Molecular

Plant, 4(2): 199-211.

Enerdata. 2012. Bilan énergétique mondial. «Quoi de neufsur la planète énergie? ». Paris;

2013.

Fan H.X., L.L. Miao, Y. Liu, H.C. Liu, Z.P. Liu. 2011. Gene cloning and characterization

of a cold-adapted beta-glucosidase belonging to glycosyl hydrolase family 1 from

a psychrotolerant bacterium Micrococcus antarcticus. Enzyme Microb. Technol.

49(1): 94-99.

Fan, G., Y. Xu, X. Zhang, S. Lei, S. Yang, and S. Pan. 2011. Characteristics of immobilized

b-glucosidase and its effect on bound volatile compounds in orange juice. Int. J.

Food Sci. Technol., 46(11): 2312–2320.

Page 271: A THESIS TITLED - prr.hec.gov.pk

References

238

Fang, S., J. Chang, Y-S. Lee, W. Guo, Y-L. Choi, and Y. Zhou. 2014. Cloning and

characterization of a new broadspecific β-glucosidase from Lactococcus sp. FSJ4.

World J. Microbiol. Biotechnol., 30(1): 213-23.

Fang, Z., W. Fang, J. Liu, Y. Hong, H. Peng, X. Zhang, B. Sun, and Y. Xiao. 2010. Cloning

and characterization of a beta-glucosidase from marine microbial metagenome with

excellent glucose tolerance. J. Microbiol. Biotechnol., 20(9): 1351-8.

Fernandes, A.N., L.H. Thomas, C.M. Altaner, P. Callow,V.T. Forsyth, D.C. Apperley, C.J.

Kennedy, M.C. Jarvis. 2011. Nanostructure of cellulose microfibrils in spruce

wood. Proceedings of the National Academy of Sciences USA, 108(47): 1195-203.

Ferreira, N.L., A. Margeot, S. Blanquet, and J.G. Berrin. 2014. Use of Cellulases from

Trichoderma reesei in the twenty first century–Part 1: Current industrial use and

future applications in the production of second generation ethanol. Biotechnol. Biol.

Trichoderma. 245-61.

Gabelsberger, J., W. Liebl, and K-H. Schleifer. 1993. Purification and properties of

recombinant β-glucosidase of the hyperthermophilic bacterium Thermotoga

maritima. Appl. Microbiol. Biotechnol., 40(1): 44–52.

Gasteiger, E., A. Gattiker, C. Hoogland, I. Ivanyi, R.D. Appel, and A. Bairoch. 2003.

ExPASy: The proteomics server for in-depth protein knowledge and analysis.

Nucleic Acids Res., 31(13): 3784-8.

Gasteiger, E., C. Hoogland, A. Gattiker, S. Duvaud, M.R. Wilkins, R.D. Appel, and A.

Bairoch. 2005. Protein Identification and Analysis Tools on the ExPASy Server.

(In) John M. Walker (ed): The Proteomics Protocols Handbook, Humana Press.

571-607.

Gitlin, I., J.D. Carbeck, and G.M. Whitesides. 2006. Why are proteins charged? Networks

of the charge-charge interactions in proteins measured by charge ladders and

capillary electrophoresis. Angew Chem Int. Ed Engl., 45(19): 3022-60.

Gloster, T.M., and G.J. Davies. 2010. Glycosidase inhibition: assessing mimicry of the

transition state. Org. Biomol. Chem., 8: 305-320.

Gloster, T.M., Madsen,R. and Davies,G.J. 2007. Structural basis for cyclophellitol

inhibition of a β-glucosidase. Org. Biomol. Chem., 5(3): 444-446.

Page 272: A THESIS TITLED - prr.hec.gov.pk

References

239

Godde, C., K. Sahm, S.J. Brouns, L.D. Kluskens, J. van der Oost, W.M. de Vos, G.

Antranikian. 2005. Cloning and expression of islandisin, a new thermostable

subtilisin from Fervidobacterium islandicum, in Escherichia coli. Appl Environ

Microbiol., 71(7): 3951-8.

Goyal, K., Jo Kim, B., Kim, J. D., Kim, Y. K., Kitaoka, M. and Hayashi, K. 2002.

Enhancement of transglycosylation activity by construction of chimeras between

mesophilic and thermophilic β-glucosidase. Arch. Biochem. Biophys., 407: 125–

134.

Goyal, K., P. Selvakumar, and K. Hayashi. 2001. Characterization of a thermostable β-

glucosidase (BglB) from Thermotoga maritima showing transglycosylation

activity. Journal of Molecular Catalysis B: Enzymatic., 15(1-3): 45-53.

Gräbnitz, F., M. Seiss, K.P. Rücknagel, and W.L. Staudenbauer. 1991. Structure of the β-

glucosidase gene bglA of Clostridium thermocellum. Sequence analysis reveals a

superfamily of cellulases and β-glycosidases including human lactase/phlorizin

hydrolase. Eur. J. Biochem., 200(2): 301–309.

Graebnitz, F., K.P. Ruecknagel, M. Seiss, and W.L. Staudenbauer. 1989. Nucleotide

sequence of the Clostridium thermocellum bgIB gene encoding thermostable beta-

glucosidase B: homology to fungal beta-glucosidases. Mol. Gen. Genet., 217(1):

70-76.

Greene et al., 2004. Growing energy. How biofuels can help end America’s oil

dependence. Nat. Res. Def. Council Rep., 1-86.

Gu, N-Y., J-L. Kim, H-J. Kim, D-J. You, H-W. Kim, and S-J. Jeon. 2009. Gene cloning

and enzymatic properties of hyperthermostable beta-glycosidase from Thermus

thermophilus HJ6. J. Biosci. Bioeng., 107(1): 21-6.

Gumerov, V.M., A.L. Rakitin, A.V. Mardanov, and N.V. Ravin. 2015. A Novel Highly

Thermostable Multifunctional Beta-Glycosidase from Crenarchaeon Acidilobus

saccharovorans. Archaea, 2015(1): 978632.

Gupta, P., S. Verma and J. Vakhlu. 2014. Comparative analysis of β-glucosidases

thermostability: Differences in amino acids composition and distribution among

mesostable and thermostable β-glucosidases J. Adv. Bioinfor. Appli. Res. 5 (3): 215-

227.

Page 273: A THESIS TITLED - prr.hec.gov.pk

References

240

Guruprasad, K., B.V.B. Reddy, M.W. Pandit. 1990. Correlation between stability of a

protein and its dipeptide composition: a novel approach for predicting in vivo

stability of a protein from its primary sequence. Protein Eng., 4(2):155-161.

Ha, C.E., and N.V. Bhagavan. 2011. Essentials of Medical Biochemistry: With Clinical

Cases. Academic Press (Elsevier Science), Burlington.

Hameed, U., I.U. Haq, and M.A. Khan. 2014. Lactose Induced expression of Thermotoga

petrophila α-amylase gene regulated by T7 promoter in E. coli CodonPlus (DE3).

Int. J. Agric. Biol., 16(4): 836‒840.

Haq, I.U., B. Muneer, Z. Hussain, M.A. Khan, S. Afzal, S. Majeed, F. Akram, and S. Akmal

2015a. Thermodynamic and saccharification analysis of cloned GH12 endo-1,4-β-

glucanase from Thermotoga petrophila in a mesophilic host. Protein Pept Lett.,

22(9): 785-794.

Haq, I.U., F. Akram, M.A. Khan, Z. Hussain, A. Nawaz, K. Iqbal, and A.J. Shah. 2015b.

CenC, a multidomain thermostable GH9 processive endoglucanase from

Clostridium thermocellum: cloning, characterization and saccharification studies.

World J. Microbiol. Biotechnol., 31(11): 1699-1710.

Haq, I.U., M.A. Khan, B. Muneer, Z. Hussain, S. Afzal, S. Majeed, N. Rashid, M.M. Javed,

and I. Ahmad. 2012b. Cloning, characterization and molecular docking of a highly

thermostable β-1,4-glucosidase from Thermotoga petrophila. Biotechnol. Lett.,

34(9): 1703-9.

Haq, I.U., Z. Hussain, M.A. Khan, B. Muneer, S. Afzal, S. Majeed, and F. Akram. 2012a.

Kinetic and thermodynamic study of cloned thermostable endo-1,4-β-xylanase

from Thermotoga petrophila in mesophilic host. Mol. Biol. Rep., 39(7): 7251-7261.

Harhangi, H.R., P.J.M. Steenbakkers, A. Akhmanova, M.S. Jetten, C. van der Drift, and

H.J. Op den Camp. 2002. A highly expressed family 1 beta-glucosidase with

transglycosylation capacity from the anaerobic fungus Piromyces sp. E2. Biochim.

Biophys. Acta, 1574(3): 293–303.

Harnpicharnchai, P., V. Champreda, W. Sornlake, and L. Eurwilaichitr. 2009. A

thermotolerant beta-glucosidase isolated from an endophytic fungi, Periconia sp.,

with a possible use for biomass conversion to sugars. Protein Expr. Purif., 67(2):

61-9.

Page 274: A THESIS TITLED - prr.hec.gov.pk

References

241

Harris, P.V., D. Welner, K.C. McFarland, E. Re, J.N. Poulsen, K. Brown, R. Salbo, H.

Ding, E. Vlasenko, S. Merino, F. Xu, J. Cherry, S. Larsen, and L.L. Leggio. 2010.

Stimulation of lignocellulosic biomass hydrolysis by proteins of glycoside

hydrolase family 61: structure and function of a large, enigmatic family.

Biochemistry, 49(15): 3305-3316.

Henrissat, B., and A. Bairoch. 1996. Updating the sequence-based classification of

glycosyl hydrolases. Biochem. J., 316 (Pt 2): 695–696.

Henrissat, B., I. Callebaut, S. Fabrega, P. Lehn, J.P. Mornon, and G. Davies. 1995.

Conserved catalytic machinery and the prediction of a common fold for several

families of glycosyl hydrolases. Proc. Natl. Acad. Sci. USA., 92(15): 7090-7094.

Himmel, M.E., and E.A. Bayer. 2009. Lignocellulose conversion to biofuels: current

challenges, global perspectives. Curr. Opin. Biotechnol., 20(3): 316-7.

Hong, J., H. Tamaki, and H. Kumagai. 2007. Cloning and functional expression of

thermostable beta-glucosidase gene from Thermoascus aurantiacus. Appl.

Microbiol. Biotechnol., 73(6): 1331-1339.

Hong, M.R., Y.S. Kim, C.S. Park, J.K. Lee, Y.S. Kim, and D.K. Oh. 2009. Characterization

of a recombinant beta-glucosidase from the thermophilic bacterium

Caldicellulosiruptor saccharolyticus. J. Biosci. Bioeng., 108(1): 36-40.

Hong, S-Y., K-M. Cho, Y-H. Kim, S-J. Hong, S-J. Cho, Y-U. Cho, H. Kim, H-D. Yun.

2006. Cloning and identification of essential residues for thermostable β-

glucosidase (BgIB) from Thermotoga maritima. J. Life Sci., 16(7): 1148-1157.

Horn, S.J., G. Vaaje-Kolstad, B. Westereng, and V.G. Eijsink. 2012. Novel enzymes for

the degradation of cellulose. Biotechnol. Biofuels, 5(1): 45.

Hu, S.C., K. Hong, Y.C. Song, J.Y. Liu, and R.X. Tan. 2009. Biotransformation of soybean

isoflavones by a marine Streptomyces sp. 060524 and cytotoxicity of the products.

W. J. Microbiol. Biotechnol., 25(1): 115-121.

Huber, R., H. Huber and K.O. Stetter. 2000. Towards the ecology of hyperthermophiles:

biotopes, new isolation strategies and novel metabolic properties. FEMS Microbiol.

Rev., 24(5): 615-23.

Ikai, A.J. 1980. Thermostability and aliphatic index of globular proteins. J. Biochem. 88(6):

1895-1898.

Page 275: A THESIS TITLED - prr.hec.gov.pk

References

242

Ikram, N., S. Naz, M.I. Rajoka, S. Sadaf, and M.W. Akhtar. 2009. Enhanced production of

subtilisin of Pyrococcus furiosus expressed in Escherichia coli using auto-inducing

medium. African J. Biotechnol., 8: 5867-5872.

Iqbal, N.M.H., I. Ahmed, A.M. Zia, and M. Irfan. 2011. Purification and characterization

of the kinetic parameters of cellulase produced from wheat straw by Trichoderma

viride under SSF and its detergent compatibility. Advanc. Bioscie. Biotechnol.,

2(3):149–156.

Irwin, D.C., S. Zhang, and D.B. Wilson. 2000. Cloning, expression and characterization of

a family 48 exocellulase, Cel48A, from Thermobifidafusca. Eur. J. Biochem., 267:

4988–4997.

Izumi, T., M.K. Piskula, S. Osawa, A. Obata, K. Tobe, M. Saito, S. Kataoka, Y. Kubota,

and M. Kikuchi. 2000. Soy isoflavone aglycones are absorbed faster and in higher

amounts than their glucosides in humans. J. Nutr., 130(7): 1695–1699.

Jabbour, D., B. Klippel, and G. Antranikian. 2012. A novel thermostable and glucose-

tolerant β-glucosidase from Fervidobacterium islandicum. Appl. Microbiol.

Biotechnol., 93(5): 1947-1956.

Jager, G., and J. Buchs. 2012. Biocatalytic conversion of lignocellulose to platform

chemicals. Biotechnol. J., 7(9): 1122–1136.

Jenkins, J., L.L. Leggio, G. Harris, and R. Pickersgill. 1995. β-Glucosidase, β-

galactosidase, family A cellulases, family F xylanases and two barley glycanases

form a superfamily of enzymes wit 8-fold β/α architecture and with two conserved

glutamates near the carboxy-terminal ends of β-strands four and seven. FEBS

Lett., 362(3): 281-285.

Jiang, C., S.X. Li, F.F. Luo, K. Jin, Q. Wang, Z.Y. Hao, L.L. Wu, G.C. Zhao, G.F. Ma,

P.H. Shen, X.L. Tang, and B. Wu. 2011. Biochemical characterization of two novel

β-glucosidase genes by metagenome expression cloning. Bioresour. Technol.,

102(3): 3272-8.

Jianzhu, M., S. Wang, F. Zhao, and J. Xu. 2013. Protein threading using context-specific

alignment potential. Bioinformatics, 29(13): i257-i265.

Page 276: A THESIS TITLED - prr.hec.gov.pk

References

243

Jun, S.Y., K.M. Park, K.W. Choi, M.K. Jang, H.Y. Kang, S.H. Lee, K.H. Park, J. Cha.

2008. Inhibitory effects of arbutin-beta-glycosides synthesized from enzymatic

transglycosylation for melanogenesis. Biotechnol Lett., 30(4):743-8.

Källberg, M., H. Wang, S. Wang, J. Peng, Z. Wang, H. Lu, and J. Xu. 2012. Template-

based protein structure modeling using the RaptorX web server. Nat. Protoc., 7(8):

1511-1522.

Källberg, M., H. Wang, S. Wang, J. Peng, Z. Wang, H. Lu, and J. Xu. 2012. Template-

based protein structure modeling using the RaptorX web server. Nature Protocols.

7: 1511-1522, 2012.

Kang, S.K., K.K. Cho, J.K. Ahn, J.D. Bok, S.H. Kang, J.H. Woo, H.G. Lee, S.K. You, and

Y.J. Choi. 2005. Three forms of thermostable lactose-hydrolase from Thermus

sp. IB 21: cloning, expression, and enzyme characterization. J. Biotechnol., 116 (4):

337-46.

Karnaouri, A., E. Topakas, T. Paschos, I. Taouki, and P. Christakopoulos. 2013. Cloning,

expression and characterization of an ethanol tolerant GH3 β glucosidase from

Myceliophthora thermophila. Peer J., 1(1): e46.

Kataeva, I. A., R.D. Seidel, A.III. Shah, L.T. West, X.L. Li, and L.G. Ljungdahl. 2002.

The fibronectin type 3-like repeat from the Clostridium thermocellum

cellobiohydrolase CbhA promotes hydrolysis of cellulose by modifying its surface.

Appl. Environ. Microbiol., 68(9): 4292-4300.

Kaur, J., B.S. Chadha, B.A. Kumar, G.S. Kaur, and H.S. Saini. 2007. Purification and

characterization of β-glucosidase from Melanocarpus sp. MTCC 3922. Elect. J.

Biotechnol., 10(2): 260-270.

Kawai, R., M. Yoshida, T. Tani, K. Igarashi, T. Ohira, H. Nagasawa, M. Samejima. 2003.

Production and characterization of recombinant Phanerochaete chrysosporium

beta-glucosidase in the methylotrophic yeast Pichia pastoris. Biosci. Biotechnol.

Biochem., 67(1): 1-7.

Kawai, T., H. Nakazawa, N. Ida, and Y. Kobayashi. 2013. A comprehensive analysis of

the effects of the main component enzymes of cellulose derived from Trichoderma

reesei on biomass saccharification. J. Ind. Microbiol. Biotechnol., 40(8): 805-10.

Page 277: A THESIS TITLED - prr.hec.gov.pk

References

244

Keerti, A. Gupta, V. Kumar, A. Dubey, and A.K. Verma. 2014. Kinetic characterization

and effect of immobilized thermostable beta-glucosidase in alginate gel Beads on

sugarcane juice. ISRN biochemistry, 2014: Article ID 178498.

Kempton, J.B., and S.G. Withers. 1992. Mechanism of Agrobacterium β-glucosidase:

kinetic studies. Biochemistry, 31(41): 9961-9969.

Kengen, S.W.M., E.J. Luesink, A.J.M. Stams, and A.J.B. Zehnder. 1993. Purification and

characterization of an extremely thermostable β-glucosidase from the

hyperthermophilic archaeon Pyrococcus furiosus. Eur. J. Biochem., 213(1): 305-

312.

Khelil, O., and B. Cheba. 2014. Thermophilic cellulolytic microorganisms from western

Algerian sources: promising isolates for cellulosic biomass recycling. Procedia

Technol., 12: 519-28.

Kibbe, W.A. 2007. OligoCalc: an online oligonucleotide properties calculator. Nucleic

Acids Res 35(Suppl 2): W43–W46. doi:10.1093/nar/gkm234.

Kiefer, F., K. Arnold, M. Künzli, L. Bordoli, and T. Schwede. 2009. The SWISS-MODEL

Repository and associated resources. Nucleic Acids Res., 37: 387-392.

Kilikian, B.V., I.D. Suarez, C.W. Liria, A.K. Gombert. 2000. Process strategies to improve

heterologous protein production in Escherchia coli under lactose and IPTG

induction. Process Biochem., 35(9): 1019–1025.

Kim, B.N., S.J. Yeom, Y.S. Kim, and D.K. Oh. 2012. Characterization of a β-glucosidase

from Sulfolobus solfataricus for isoflavone glycosides. Biotechnol. Lett., 34(1):

125-9.

Kim, H.J., A-R. Park, J-K. Lee, and D.K. Oh. 2009. Characterization of an acid-labile,

thermostable β-glycosidase from Thermoplasma acidophilum. Biotechnol. Lett.,

31(9): 1457-62.

Kim, J.S., Y.Y. Lee and R.W. Torget. 2001. Cellulose hydrolysis under extremely low

sulfuric acid and high-temperature conditions. Appl. Biochem. Biotechnol., 91-93:

331–340.

Kim, S.J., C.M. Lee, M.Y. Kim, Y.S. Yeo, S.H. Yoon, H.C. Kang, and B.S. Koo. 2007.

Screening and characterization of an enzyme with beta-glucosidase activity from

environmental DNA. J. Microbiol. Biotechnol., 17(6): 905-12.

Page 278: A THESIS TITLED - prr.hec.gov.pk

References

245

Kim, Y.S., S.J. Yeom, and D.K. Oh. 2011. Characterization of a GH3 family β-glucosidase

from Dictyoglomus turgidum and its application to the hydrolysis of isoflavone

glycosides in spent coffee grounds. J. Agric. Food Chem., 59(21): 11812-8.

Klose, H., J. Röder, M. Girfoglio, R. Fischer, and U. Commandeur. 2012.

Hyperthermophilic endoglucanase for in planta lignocellulose conversion.

Biotechnol. Biofuels, 5(1): 63.

Koppram, R., E. Tomás-Pejó, C. Xiros, L. Olsson. 2014. Lignocellulosic ethanol

production at high-gravity: challenges and perspectives. Trends Biotechnol.,

32(1):46-53.

Krisch, J., M. Takó, T. Papp and C. Vágvölgyi. 2010. Characteristics and potential use of

β-glucosidases from Zygomycetes. Current Research; Technology and Education

Topics in Applied Microbiology and Microbial Biotechnology, Edition:

MICROBIOLOGY BOOK SERIES - Number 2, Publisher: Formatex Research

Center, Editors: Méndez-Vilas A, 891-896.

Kuhad, R.C., D. Deswal, S. Sharma, A. Bhattacharya, K.K. Jain, A. Kaur, B.I. Pletschke,

A. Singh, M. Karp. 2016. Revisiting cellulase production and redefining current

strategies based on major challenges. Renewable and Sustainable Energy Reviews.

55: 249-272.

Kuhad, R.C., R. Gupta, and A. Singh. 2011. Microbial cellulases and their industrial

applications. Enzyme Res., 2011: 280696.

Kumar, L., G. Awasthi, and B. Singh. 2011. Extremophiles: A noval source of industrially

important enzymes. Biotechnol., 10(2): 121-35.

Kuo, L.C., and K-T. Lee. 2008. Cloning, expression, and characterization of two β-

glucosidases from isoflavone glycoside-hydrolyzing Bacillus subtilis natto. J.

Agric. Food Chem., 56(1): 119-25.

Laidler, K.J., and B.F. Peterman. 2009. Temperature effects on enzymes kinetics. In D.

Purich (ed.), contemporary enzymes kinetics and mechanism, pp: 177-197.

Academic Press.

Lamers, P., K. McCormick, J.N. Hilbert. 2008. The emerging liquid biofuel market in

Argentina: implications for domestic demand and international trade. Energy

Policy. 36(4): 1479-1490.

Page 279: A THESIS TITLED - prr.hec.gov.pk

References

246

Laskowski, R.A. 2009. PDBsum new things. Nucleic Acids Res., 37: D355-D359.

Li, D., X. Li, W. Dang, P.L. Tran, S-H. Park, B-C. Oh, W-S. Hong, J.S. Lee, and K-H.

Park. 2013. Characterization and application of an acidophilic and thermostable β-

glucosidase from Thermofilum pendens. J. Biosci. Bioeng., 115(5): 490-6.

Li, G., Y. Jiang, X.J. Fan, and Y.H. Liu. 2012. Molecular cloning and characterization of

a novel β-glucosidase with high hydrolyzing ability for soybean isoflavone

glycosides and glucose-tolerance from soil metagenomic library. Bioresour

Technol., 123: 15-22.

Li, W.F., X.X. Zhou, and P. Lu. 2005. Structural features of thermozymes. Review article.

Biotechnol. Adv., 23(4):271-281.

Li, Y-K., and J-A. Lee. 1999. Cloning and expression of β-glucosidase

from Flavobacterium meningosepticum: A new member of family B β-glucosidase.

Enzyme Microbial. Technol., 24(3): 144-150.

Lieberman, R.L., B.A. Wustman, P. Huertas, Jr.A.C. Powe, C.W. Pine, R. Khanna, M.G.

Schlossmacher, D. Ringe, and G.A. Petsko. 2007. Structure of acid betaglucosidase

with pharmacological chaperone provides insight into Gaucher disease. Nat. Chem.

Biol., 3(2): 101–107.

Litzinger, S., S. Fischer, P. Polzer, K. Diederichs, W. Welte, C. Mayer. 2010. Structural

and kinetic analysis of Bacillus subtilis N-acetylglucosaminidase reveals a unique

Asp-His dyad mechanism. J. Biol. Chem., 285(46):35675-35684.

Liu, D., R. Zhang, X. Yang, Z. Zhang, S. Song, Y. Miao, and Q. Shen. 2012.

Characterization of a thermostable β-glucosidase from Aspergillus fumigatus Z5,

and its functional expression in Pichia pastoris X33. Microb. Cell Fact., 17(11): 25.

Lombard, V., H. Golaconda Ramulu, E. Drula, P.M. Coutinho, and B. Henrissat. 2014. The

carbohydrate-active enzyme database (CAZy) in 2013. Nucleic Acids Res., 42:

D490-D495.

Long, L., H. Shi, X. Li, Y. Zhang, J. Hu, and F. Wang. 2016. Cloning, Purification, and

Characterization of a Thermostable β-Glucosidase from Thermotoga thermarum

DSM 5069. BioResource, 11(2): 3165-3177.

Page 280: A THESIS TITLED - prr.hec.gov.pk

References

247

Lu, J., L. Du, Y. Wei, Y. Hu, and R. Huang. 2013. Expression and characterization of a

novel highly glucose-tolerant β-glucosidase from a soil metagenome. Acta.

Biochim. Biophys. Sin (Shanghai), 45(8): 664-73.

Lynd, L.R., P.J. Weimer, W.H. Van-Zyl, I.S. Pretorius. 2002. Microbial cellulose

utilization: fundamentals and biotechnology. Microbiol. Mol. Bio Rev., 66(3): 506-

77.

Maity, S.K. 2015. Opportunities, recent trends and challenges of integrated Biorefinery:

Part I. Renew. Sustainable Energy Rev., 43: 1427–1445.

Maki, M., K.T. Leung, W. Qin. 2009. The prospects of cellulase-producing bacteria for the

bioconversion of lignocellulosic biomass. Int. J. Biol. Sci., 5(5): 500-516.

Marchler-Bauer, A., M.K. Derbyshire, N.R. Gonzales, S. Lu., F. Chitsaz, L.Y. Geer, R.C.

Geer, J. He, M. Gwadz, D.I. Hurwitz, C.J. Lanczycki, F. Lu, G.H. Marchler, J.S.

Song, N. Thanki, Z. Wang, R.A. Yamashita, D. Zhang, C. Zheng, and S.H. Bryant.

2015. CDD: NCBI’s conserved domain database. Nucleic Acids Res., 43: 222-226.

Marín, E., L. Sánchez, M.D. Pérez, P. Puyol, M. Calvo. 2003. Effect of heat treatment on

bovine lactoperoxidase activity in skim milk: Kinetic and thermodynamic analysis.

J. Food Sci., 68(1): 89–93.

Marotti, I., A. Bonetti, B. Biavati, P. Catizone, and G. Dinelli. 2007. Biotransformation of

common bean (Phaseolus vulgaris L.) flavonoid glycosides by bifidobacterium

species from human intestinal origin. J. Agric. Food Chem., 55(10): 3913-3919.

Matthews, B.W., H. Nicholson, W.J. Becktel. 1987. Enhanced protein thermostability from

site-directed mutations that decrease the entropy of unfolding. Proc. Natl. Acad.

Sci. USA., 84(19) :6663–6667.

Mayer, S., S. Junne, K. Ukkonen, J. Glazyrina, F. Glauche, P. Neubauer, and A. Vasala.

2014. Lactose auto-induction with enzymatic glucose release: characterization of

the cultivation system in bioreactor. Protein Expr. Purif., 94: 67-72.

McCarter, J.D., and S.G. Withers. 1994. Mechanisms of enzymatic glycoside hydrolysis.

Curr. Opin. Struct. Biol., 4(6): 885-92.

Mehmood, M.A., I. Shahid, K. Hussain, F. Latif, and M.I. Rajoka. 2014. Thermodynamic

properties of the β-glucosidase from Thermotoga maritima extend the upper limit

of thermophilicity. Protein Pept. Lett., 21 (12): 1282-8.

Page 281: A THESIS TITLED - prr.hec.gov.pk

References

248

Mihasan, M., E. Ungureanu, and V. Artenie. 2007. Optimum parameters for over

expression of recombinant protein from tacpromotors on auto-inducible medium.

Romanian Biotechnol. Letters, 12: 3473.

Miller, G.L. 1959. Use of dinitrosalicylic acid reagent for determination of reducing sugar.

Anal. Chem., 31(3): 426–428.

Mohanram, S., D. Amat, J. Choudhary, A. Arora, and L. Nain. 2013. Novel perspectives

for evolving enzyme cocktails for lignocellulose hydrolysis in biorefineries.

Sustainable Chemical Processes, 1: 15.

Moracci, M., C. Luisa, C. Maria, and R. Mose. 1996. Identification of two glutamic acid

residues essential for catalysis in the β-glucosidase from the thermoacidophilic

archaeon Sulfolobus solfataricus. Protein Eng., 9(12): 1191-1195.

Nam, E.S., M.S. Kim, H.B. Lee, and J.K. Ahn. 2010. Beta-glycosidase of Thermus

thermophilus KNOUC202: gene and biochemical properties of the enzyme

expressed in Escherichia coli. Prikl. Biokhim. Mikrobiol., 46(5): 562-71.

Naz, S., N. Ikram, M.I. Rajoka, S. Sadaf, and M.W. Akhtar. 2010. Enhanced production

and characterization of a β-Glucosidase from Bacillus halodurans expressed in

Escherichia coli. Biochemistry (Mosc), 75(4): 513-25.

Niehaus, F., C. Bertoldo, M. Kähler, G. Antranikian. 1999. Extremophiles as a source of

novel enzymes for industrial application. Appl. Microbiol. Biotechnol., 51(6):711-

29.

Nijikken, Y., T. Tsukada, K. Igarashi, M. Samejima, T. Wakagi, H. Shoun, and S.

Fushinobu. 2007. Crystal structure of intracellular family 1 β-glucosidase BGL1A

from the basidiomycete Phanerochaete chrysosporium. FEBS Lett., 581(7): 1514-

1520.

Nunoura, N., K. Ohdan, T. Yano, K. Yamamoto, H. Kumagai. 1996. Purification and

characterization of beta-D-glucosidase (beta-D-fucosidase) from Bifidobacterium

breve clb acclimated to cellobiose. Biosci. Biotechnol. Biochem., 60(2): 188-93.

Oganesyan, N., I. Ankoudinova, S.H. Kim, and R. Kim. 2007. Effect of osmotic stress and

heat shock in recombinant protein overexpression and crystallization. Protein Expr.

Purif., 52(2): 280–285.

Page 282: A THESIS TITLED - prr.hec.gov.pk

References

249

Opassiri, R., B. Pomthong, T. Akiyama, M. Nakphaichit, T. Onkoksoong, M. Ketudat

Cairns, and J.R. Ketudat Cairns. 2007. A stress-induced rice (Oryza sativa L.) beta-

glucosidase represents a new subfamily of glycosyl hydrolase family 5 containing

a fascin-like domain. Biochem. J., 408(2): 241-249.

Opassiri, R., Y. Hua, O. Wara-Aswapati, T. Akiyama, J. Svasti, A. Esen, and J.R.K. Cairns.

2004. Beta-glucosidase, exo-beta-glucanase and pyridoxine transglucosylase

activities of rice BGlu1. Biochem. J., 379(1): 125–131.

Park, N.Y., J. Cha, D.O. Kim, and C.S. Park. 2007. Enzymatic characterization and

substrate specificity of thermostable beta-glycosidase from hyperthermophilic

archaea, Sulfolobus shibatae, expressed in E. coli. J. Microbiol. Biotechnol., 17(3):

454-60.

Park, T.H., K.W. Choi, C.S. Park, S.B. Lee, H.Y. Kang, K.J. Shon, J.S. Park, and J. Cha.

2005. Substrate specificity and transglycosylation catalyzed by a thermostable beta-

glucosidase from marine hyperthermophile Thermotoga neapolitana. Appl.

Microbiol. Biotechnol., 69(4): 411-22.

Parry, N.J., D.E. Beever, E. Owen, I. Vandenberghe, J.V. Beeumen and M.K. Bhat. 2001.

Biochemical characterization and mechanism of action of a thermostable β-

glucosidase purified from Thermoascus aurantiacus. Biochem. J., 353(1): 117–

127.

Pei, J., Q. Pang, L. Zhao, S. Fan, and H. Shi. 2012. Thermoanaerobacterium

thermosaccharolyticum β-glucosidase: a glucose-tolerant enzyme with high

specific activity for cellobiose. Biotechnol. Biofuels, 5(1): 31.

Peng, X., H. Su, S. Mi, and Y. Han. 2016. A multifunctional thermophilic glycoside

hydrolase from Caldicellulosiruptor owensensis with potential applications in

production of biofuels and biochemicals. Biotechnol. Biofuels, 9(1): 98.

Pereira, S.C., L. Maehara, C.M. Machado, and C.S. Farinas. 2015. 2G ethanol from the

whole sugarcane lignocellulosic biomass. Biotechnol. Biofuels, 8: 44.

Pisani, F.M., R. Rella, C.A. Raia, C. Rozzo, R. Nucci, A. Gambacorta, M. De Rosa, and

M. Rossi. 1990. Thermostable β-galactosidase from the archaebacterium

Sulfolobus solfataricus Purification and properties. Eur. J. Biochem., 187(2): 321 -

328.

Page 283: A THESIS TITLED - prr.hec.gov.pk

References

250

Plant, A.R., J.E. Oliver, M.L. Patchett, R.M. Daniel, and H.W. Morgan. 1988. Stability and

substrate specificity of a β-glucosidase from the thermophilic bacterium Tp8 cloned

into Escherichia coli. Arch. Biochem. Biophys., 262(1): 181-188.

Pozzo, T., J.L. Pasten, E.N. Karlsson and D.T. Logan. 2010. Structural and functional

analyses of β-glucosidase 3B from Thermotoga neapolitana: A thermostable three-

domain representative of glycoside hydrolase 3. J. Mol. Biol., 397(3): 724-739.

Raghuwanshi, S., D. Deswal, M. Karp, and R.C. Kuhad. 2014. Bioprocessing of enhanced

cellulase production from a mutant of Trichoderma asperellum RCK2011 and its

application in hydrolysis of cellulose. Fuel, 124: 183-189.

Rajoka, M.I., S. Idrees, U.A. Ashfaq, B. Ehsan, and A. Haq. 2015. Determination of

substrate specificities against β-glucosidase A (BglA) from Thermotoga maritime:

a molecular docking approach. J. Microbiol. Biotechnol., 25(1): 44-9.

Ramani, G., B. Meera, C. Vanitha, J. Rajendhran, and P. Gunasekaran. 2015. Molecular

cloning and expression of thermostable glucose tolerant β-glucosidase of

Penicillium funiculosum NCL1 in Pichia pastoris and its characterization. J. Ind.

Microbiol. Biotechnol., 42(4): 553-65.

Ramírez-Escudero, M., M.V. Del-Pozo, J. Marín-Navarro, B. González, P.N. Golyshin, J,

Polaina, M. Ferrer, J. Sanz-Aparicio. 2016. Structural and Functional

Characterization of a Ruminal β-Glycosidase Defines a Novel Subfamily of

Glycoside Hydrolase Family 3 with Permuted Domain Topology. J. Biol. Chem.,

291(46): 24200-24214.

Riou, C., J.M. Salmon, M.J. Vallier, Z. Günata, and P. Barre. 1998. Purification,

characterization, and substrate specificity of a novel highly glucose-tolerant β-

glucosidase from Aspergillus oryzae. Appl. Environ. Microbiol., 64: 3607-3614.

Rojas, J., M. Bedoya, and Y. Ciro. 2015. "Cellulose-Fundamental Aspects and Current

Trends", Chapter No. 08, Current Trends in the Production of Cellulose

Nanoparticles and Nanocomposites for Biomedical Applications. Academic Press:

10.5772/61334.

Rosano, G.L., E.A. Ceccarelli. 2014. Recombinant protein expression in Escherichia coli:

advances and challenges. Front. Microbiol., 5:172.

Page 284: A THESIS TITLED - prr.hec.gov.pk

References

251

Ruttersmith, L.D., and R.M. Daniel. 1993. Thermostable beta-glucosidase and beta-

xylosidase from Thermotoga sp. strain FjSS3-B.1. Biochim. Biophys. Acta.,

1156(2): 167-72.

Sambrook, J., Russell, D.W. 2001. Molecular cloning: a laboratory manual. Cold Spring

Harbor, New York.

Sanchez-Ruiz, J.M. 2010. Protein kinetic stability. Biophys. Chem., 148(1-3): 1–15.

Saratale, G. D., and S. E. Oh. 2012. Lignocellulosics to ethanol: the future of the chemical

and energy industry. Afr. J. Biotechnol. 11(5):1002-1013.

Schneidman-Duhovny, D., Y. Inbar, R. Nussinov, and H.J. Wolfson. 2005. PatchDock and

SymmDock: servers for rigid and symmetric docking. Nucleic Acids Res., 33,

W363-367.

Schroder, C., S. Elleuche, S. Blank, and G. Antranikian. 2014. Characterization of a heat-

active archaeal β-glucosidase from a hydrothermal spring metagenome. Enzyme

Microb. Technol., 57: 48-54.

Shi, H., Y. Zhang, H. Zhong, Y. Huang, X. Li, and F. Wang. 2014. Cloning, over-

expression and characterization of a thermo-tolerant xylanase from Thermotoga

thermarum. Biotechnol. letters, 36(3): 587-593.

Shi, H., Y. Zhang, X. Li, Y. Huang, L. Wang, Y. Wang, H. Ding, and F. Wang. 2013. A

novel highly thermostable xylanase stimulated by Ca2+ from Thermotoga

thermarum: cloning, expression and characterization. Biotechnol. Biofuels, 6(1):

26.

Shin, K.C., and D.K. Oh. 2014. Characterization of a novel recombinant β-glucosidase

from Sphingopyxis alaskensis that specifically hydrolyzes the outer glucose at the

C-3 position in protopanaxadiol-type ginsenosides. J. Biotechnol., 172: 30-7.

Singh, A., and K. Hayashi. 1995. Construction of chimeric beta-glucosidases with

improved enzymatic properties. J. Biol. Chem., 270(37): 21928-33.

Singh, G., A.K. Verma, and V. Kumar. 2016. Catalytic properties, functional attributes and

industrial applications of β-glucosidases. 3 Biotech., 6(1): 3.

Singhania, R.R., A.K. Patel, R.K. Sukumaran, C. Larroche, and A. Pandey. 2012. Role and

significance of beta-glucosidases in the hydrolysis of cellulose for bioethanol

production. Bioresour. Technol., 127: 500-7.

Page 285: A THESIS TITLED - prr.hec.gov.pk

References

252

Sohel, M.I., and M.W. Jack. 2012. Thermodynamic analysis of a high-yield biochemical

process for biofuel production. Bioresour Technol., 124: 406-412.

Sørensen, A., M. Lübeck, P.S. Lübeck, and B.K. Ahring. 2013. Fungal beta-glucosidases:

a bottleneck in industrial use of lignocellulosic materials. Biomolecules, 3(3): 612-

631.

Stoker, H.S. 2008. General, Organic, and Biological Chemistry. 5th Ed. Brooks/Cole-

Cengage Learning, Belmont, CA.

Street, I.P., J.B. Kempton, and S.G. Withers. 1992. Inactivation of a β-glucosidase through

the accumulation of a stable 2-deoxy-2-fluoro-alpha-D-glucopyranosyl-enzyme

intermediate: a detailed investigation. Biochemistry, 31(41): 9970-9978.

Studier, F.W. 2005. Protein production by auto-induction in high-density shaking cultures.

Protein Expr. Purif., 41: 207-234.

Su, E., T. Xia, L. Gao, Q. Dai, and Z. Zhang. 2010. Immobilization of beta-glucosidase

and its aroma-increasing effect on tea beverage. Food Bioprod. Process, 88(2): 83–

89.

Su, L., Y. Huang, and J. Wu. 2015. Enhanced production of recombinant Escherichia coli

glutamate decarboxylase through optimization of induction strategy and addition

of pyridoxine. Bioresour. Technol., 198: 63-69.

Suhre, K., J.M. Claverie. 2003. Genomic correlates of hyperthermostability, an update. J.

Biol. Chem., 278: 17198-17202.

Svetlitchnyi, V.A., O. Kensch, D.A. Falkenhan, S.G. Korseska, N. Lippert, M. Prinz, J.

Sassi, A. Schickor, and S. Curvers. 2013. Single-step ethanol production from

lignocellulose using novel extremely thermophilic bacteria. Biotechnol. Biofuels,

6(1): 31.

Tajima, K., K. Nakajima, H. Yamashita, T. Shiba, M. Munekata, and M. Takai. 2001.

Cloning and sequencing of the Beta-glucosidase gene from Acetobacter xylinum

ATCC 23769. DNA Res., 8(6): 263-269.

Takahata, Y., M. Nishijima, T. Hoaki and T. Maruyama. 2001. Thermotoga petrophila sp.

nov. and Thermotoga naphthophila sp. nov., two hyperthermophilic bacteria from

the Kubiki oil reservoir in Niigata, Japan. Int. Jour. Syst. Evol. Microbiol., 51(5):

1901-9.

Page 286: A THESIS TITLED - prr.hec.gov.pk

References

253

Tang, Z., S. Liu, H. Jing, R. Sun, M. Liu, H. Chen, Q. Wu, and X. Han. 2014. Cloning and

expression of Aspergillus oryzae β-glucosidase in Pichia pastoris. Mol. Biol.

Rep., 41(11): 7567-73.

Tayefi-Nasrabadi, H., R. Asadpour. 2008. Effect of heat treatment on buffalo (Bubalus

bubalis) lactoperoxidase activity in raw milk. J. Biol. Sci., 8(8):1310-1315.

Teugjas, H., and P. Väljamäe. 2013. Selecting β-glucosidases to support cellulases in

cellulose saccharification. Biotechnol. Biofuels, 6(1):105.

Thongpoo, P., L.S. McKee, A.C. Araújo, P.T. Kongsaeree, and H. Brumer. 2013.

Identification of the acid/base catalyst of a glycoside hydrolase family 3 (GH3) β-

glucosidase from Aspergillus niger ASKU28. Biochim. Biophys. Acta, 1830(3):

2739-49.

Tomme, P., R.A. Warren, and N.R. Gilkes. 1995. Cellulose Hydrolysis by Bacteria and

Fungi. Adv. Microb. Physiol., 37: 1-81.

Tran, T.T., G. Mamo, B. Mattiasson, and H.R. Kaul. 2010. A thermostable phytase from

Bacillus sp. MD2: cloning, expression and high-level production in Escherichia

coli. J. Ind. Microbiol. Biotechnol., 37: 279-287.

Tribolo, S., J.G. Berrin, P.A. Kroon, M. Czjzek, and N. Juge. 2007. The crystal structure

of human cytosolic β-glucosidase unravels the substrate aglycone specificity of a

family 1 glycoside hydrolase. J. Mol. Biol., 370(5): 964-975.

Turner, C., P. Turner, G. Jacobson, K. Almgren, M. Waldebäck, P. Sjöberg, E.N. Karlsson,

and K.E. Markides. 2006. Subcritical water extraction and β-glucosidasecatalyzed

hydrolysis of quercetin in onion waste. Green Chem., 8(11): 949-959.

Turner, P., D. Sevenson, P. Adlercreutz, E.N. Karlsson. 2007. A novel variant of

Thermotoga neapolitana β-glucosidase B is an efficient catalyst for the synthesis

of alkyl glucosides by transglycosylation. J. Biotechnol., 130(1): 67-74.

Väljamäe, P., G. Pettersson, and G. Johansson. 2001. Mechanism of substrate inhibition in

cellulose synergistic degradation. Eur. J. Biochem., 268: 4520-4526.

Vallmitjana, M., F-N. Mario, P. Raquel, A. Mireia, A. Cristina, Q. Enrique, P.

Antoni, and P-P. Josep-Anton. 2001. Mechanism of the family 1 β-glucosidase

from Streptomyces sp: Catalytic residues and kinetic studies. Biochem., 40(20):

5975-5982.

Page 287: A THESIS TITLED - prr.hec.gov.pk

References

254

VanFossen, A.L., D.L. Lewis, J.D. Nichols, and R.M. Kelly. 2008. Polysaccharide

degradation and synthesis by extremely thermophilic anaerobes. Ann. NY. Acad.

Sci., 1125: 322-337.

Vanitha, M.C., B. Meera, G. Ramani, M. Rao, S. Laxman, and P. Gunasekaran. 2011.

Molecular cloning and expression of a family 7 cellobiohydrolase gene CBH1 from

Penicillium funiculosum NCL1. Int. J. Microbiol. Res., 3(2): 97-107.

Vincze, T., J. Posfai, and R.J. Roberts. 2003. NEBcutter: A program to cleave DNA with

restriction enzymes. Nucleic Acids Res., 31(13): 3688–3691.

Volkin, D.B., A.M. Klibanov. 1989. Minimizing protein inactivation In: CreightonTE,

editors. Protein Function. A practical approach. Oxford: IRL press. pp. 1–24.

Voorhorst, W.G., R.I. Eggen, E.J. Luesink, and W.M. de-Vos. 1995. Characterization of

the celB gene coding for beta-glucosidase from the hyperthermophilic archaeon

Pyrococcus furiosus and its expression and site-directed mutation in Escherichia

coli. J. Bacteriol., 177(24): 7105-11.

Vuong, T.V., and D.B. Wilson. 2010. Glycoside hydrolases: catalytic base/nucleophile

diversity. Biotechnol. Bioeng., 107: 195-205.

Wang, M., G.L. Lai, Y. Nie, S. Geng, L. Liu, B. Zhu, Z. Shi, X.L. Wu. 2015. Synergistic

function of four novel thermostable glycoside hydrolases from a long-term enriched

thermophilic methanogenic digester. Front Microbiol., 6:509.

Wang, Q., D. Trimbur, R. Graham, R.A.J. Warren, and S.G. Withers. 1995. Identification

of the acid/base catalyst in Agrobacterium faecalis beta-glucosidase by kinetic

analysis of mutants. Biochem., 34(44): 14554-14562.

Wang, Q., C. Qian, X.Z. Zhang, N. Liu, X. Yan, and Z. Zhou. 2012. Characterization of a

novel thermostable β-glucosidase from a metagenomic library of termite gut.

Enzyme Microb. Technol., 51(6-7): 319-24.

Widdel, F., G. Kohring, F. Mayer. 1983. Studies in dissimilatory sulfate-reducing bacteria

that decompose fatty acids. III. Characterization of the filamentous gliding

Desulfonema limicola gen. nov. sp. nov., and Desulfonema magnum sp. nov. Arch.

Microbiol., 134: 286-294.

Wierenga, R.K. 2001. The TIM-barrel fold: a versatile framework for efficient enzymes.

FEBS Lett., 492(3): 193-8.

Page 288: A THESIS TITLED - prr.hec.gov.pk

References

255

Withers, S.G., K. Rupitz, D. Trimbur, and R.A. Warren. 1992. Mechanistic consequences

of mutation of the active site nucleophile Glu 358 in Agrobacterium β-glucosidase.

Biochem., 31(41): 9979-9985.

Woodley, J. M. 2013. Protein engineering of enzymes for process applications. Curr. Opin.

Chem. Biol., 17(2): 310-316.

Xiangyuan, H., Z. Shuzheng, and Y. Shoujun. 2001. Cloning and expression of

thermostable beta-glycosidase gene from Thermus nonproteolyticus HG102 and

characterization of recombinant enzyme. Appl. Biochem. Biotechnol., 94(3): 243-

55.

Xie, J., D. Zhao, L. Zhao, J. Pei, W. Xiao, G. Ding, and Z. Wang. 2015. Overexpression

and characterization of a Ca2+ activated thermostable β-glucosidase with high

ginsenoside Rb1 to ginsenoside 20(S)-Rg3 bioconversion productivity. J. Ind.

Microbiol. Biotechnol., 42(6): 839-50.

Xu, R., F. Teng, C. Zhang, and D. Li. 2011. Cloning of a gene encoding β-glucosidase

from Chaetomium thermophilum CT2 and its expression in Pichia pastoris. J. Mol.

Microbiol. Biotechnol., 20(1): 16-23.

Xue, Y., X. Song, and J. Yu. 2009. Overexpression of β-glucosidase from Thermotoga

maritima for the production of highly purified aglycone isoflavones from soy flour.

World J. Microbiol. Biotechnol., 25 (12): 2165-72.

Yan, J., S.F. Zhao, Y.F Mao, and Y.H. Luo. 2004. Effects of lactose as an inducer on

expression of Helicobacter pylori rUreB and rHpaA, and Escherichia coli

rLTKA63 and rLTB. World J. Gastroenterol., 10: 1755-1758.

Yan, T.R., and C.L. Lin. 1997. Purification and characterization of a glucose-tolerant β-

glucosidase from Aspergillus niger CCRC 31494. Biosci. Biotech. Biochem., 61(6):

965–970.

Yang, F., X. Yang, Z. Li, C. Du, J. Wang, and S. Li. 2015. Overexpression and

characterization of a glucose-tolerant β-glucosidase from T. aotearoense with high

specific activity for cellobiose. Appl. Microbiol. Biotechnol., 99(21): 8903-15.

Yang, X., R. Ma, P. Shi, H. Huang, Y. Bai, Y. Wang, P. Yang, Y. Fan, and B. Yao. 2014.

Molecular characterization of a highly-active thermophilic β-glucosidase from

Page 289: A THESIS TITLED - prr.hec.gov.pk

References

256

Neosartorya fischeri P1 and its application in the hydrolysis of soybean isoflavone

glycosides. PLoS One, 9(9).

Yennamalli, R.M., A.J. Rader, A.J. Kenny, J.D. Wolt, T.Z. Sen. 2013. Endoglucanases:

insights into thermostability for biofuel applications. Biotechnol Biofuels. 6(1):136.

Yeoman, C.J., Y. Han, D. Dodd, C.M. Schroeder, R.I. Mackie, and I.K. Cann. 2010.

Thermostable enzymes as biocatalysts in the biofuel industry. Adv. Appl.

Microbiol., 70: 1-55.

Yoon, J.J., K-Y. Kim, and C-J. Cha. 2008. Purification and characterization of

thermostable beta-glucosidase from the brown-rot basidiomycete Fomitopsis

palustris grown on microcrystalline cellulose. J. Microbiol., 46(1): 51-55.

Zafar, M., S. Ahmed, M.I. Khan and A. Jamil. 2014. Recombinant expression and

characterization of a novel endoglucanase from Bacillus subtilis in Escherichia

coli. Mol. Biol. Rep., 41(5): 3295–3302.

Zanoelo, F.F., M.L. Polizeli, H.F. Terenzi, and J.A. Jorge. 2004. Beta-glucosidase activity

from the thermophilic fungus Scytalidium thermophilum is stimulated by glucose

and xylose. FEMS Microbiol. Lett., 240(2): 137-43.

Zhang, Q., W. Zhang, C. Lin, X. Xu, and Z. Shen. 2012. Expression of an Acidothermus

cellulolyticus endoglucanase in transgenic rice seeds. Protein Expr. Purif. 82(2):

279-83.

Zhang, Y-H.P., S-Y. Ding, J. R. Mielenz, J-B. Cui, R.T. Elander, M. Laser, M.E. Himmel,

J.R. McMillan, L.R. Lynd. 2007. Fractionating Recalcitrant Lignocellulose at

Modest Reaction Conditions. Biotechnol. Bioeng., 97(2): 214-223.

Zhao, J., C. Guo, C. Tian, and Y. Ma. 2015. Heterologous Expression and Characterization

of a GH3 β-glucosidase from Thermophilic Fungi Myceliophthora thermophila in

Pichia pastoris. Appl. Biochem. Biotechnol., 177(2): 511-27.

Zhao, L., J. Xie, X. Zhang, F. Cao, and J. Pei. 2013. Overexpression and characterization

of a glucose-tolerant β-glucosidase from Thermotoga thermarum DSM 5069T with

high catalytic efficiency of ginsenoside Rb1 to Rd. J. of Mol. Catalysis B:

Enzymatic, 95: 62-69.

Zou, Z.Z., H.L. Yu, C.X. Li, X.W. Zhou, C. Hayashi, J. Sun, B.H. Liu, T. Imanaka,

and J.H. Xu. 2012. A new thermostable β-glucosidase mined from Dictyoglomus

Page 290: A THESIS TITLED - prr.hec.gov.pk

References

257

thermophilum: properties and performance in octyl glucoside synthesis at high

temperatures. Bioresour. Technol., 118: 425-430.

Zverlov, V.V., I.Y. Volkov, T.V. Velikodvorskaya, and W.H. Schwarz. 1997. Thermotoga

neapolitana bglB gene, upstream of lamA, encodes a highly thermostable beta-

glucosidase that is a laminaribiase. Microbiology, 143(11): 3537-42.


Recommended