+ All Categories
Home > Documents > ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

Date post: 14-Feb-2022
Category:
Upload: others
View: 6 times
Download: 0 times
Share this document with a friend
203
ﺑﺳﻡ ﷲ ﺍﻟﺭﺣﻣﺎﻥ ﺍﻟﺭﺣﻳﻡNEW INHIBITORS OF ANGIOTENSIN CONVERTING ENZYME FROM PLANT AND SYNTHETIC ORIGIN BY MASS SPECTROMETRIC SCREENING ASSAY Thesis submitted for the partial fulfillment of the degree of Doctor of Philosophy in Chemistry By MUHAMMAD SALMAN BHATTI H.E.J. RESEARCH INSTITUTE OF CHEMISTRY International Center for Chemical and Biological Sciences University of Karachi, Karachi-75270, Pakistan 2019
Transcript
Page 1: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

بسم الله الرحمان الرحيم

NEW INHIBITORS OF ANGIOTENSIN

CONVERTING ENZYME FROM PLANT AND

SYNTHETIC ORIGIN BY MASS SPECTROMETRIC

SCREENING ASSAY

Thesis submitted for the partial fulfillment of the degree of

Doctor of Philosophy

in

Chemistry

By

MUHAMMAD SALMAN BHATTI

H.E.J. RESEARCH INSTITUTE OF CHEMISTRY

International Center for Chemical and Biological Sciences

University of Karachi,

Karachi-75270, Pakistan

2019

Page 2: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

II

Wxw|vtàxw gÉ

`ç ctÜxÇàá

Page 3: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

III

Thesis Certificate

This is to certify that the thesis entitled, “New Inhibitors of Angiotensin

Converting Enzyme from Plant and Synthetic Origin by Mass Spectrometric

Screening Assay” has been submitted to the Advanced Studies Research Board

(ASRB), University of Karachi by Mr. Muhammad Salman Bhatti s/o Mr.

Fakhruddin Bhatti for the award of the degree, Doctor of Philosophy (Ph.D.) in

the discipline of Chemistry. The research has been carried out under my

supervision and I certify that the work submitted herein is original and not

plagiarized from any other source. Neither the thesis nor the work contained

therein has been previously submitted to any institution for a degree.

___________________________________

Dr. Syed Ghulam Musharraf

Professor H.E.J. Research Institute of Chemistry International Center for Chemical and Biological Sciences (ICCBS) University of Karachi, Karachi-75270, Pakistan

Page 4: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

IV

Contents

Acknowledgements ........................................................................................ VIII

Personal Introduction ....................................................................................... X

List of Publications .......................................................................................... XI

List of Figures ................................................................................................. XII

List of Tables ................................................................................................. XV

List of Abbreviations ...................................................................................... XVI

Summary .............................................................................................. XVIII

XIX ................................................................................................ خلاصہ

Chapter 1: Pharmacological Importance of Enzymes and

Angiotensin Converting Enzyme (ACE) ...................................... 1

1.1 Enzymes in Life: Brief History ................................................................... 2

1.2 Enzyme Catalysis ..................................................................................... 2

1.3 Αngiotensin Converting Εnzyme (ΑCΕ) .................................................... 3

1.3.1 Renin Angiotensin Aldosterone System (RAAS) ........................... 4

1.3.2 Problems Associated with Over Expression of ACE and

RAAS ............................................................................................ 6

1.4 Enzyme Inhibition ..................................................................................... 8

1.4.1 Reversible Inhibition ...................................................................... 9

1.4.2 Irreversible Inhibition ................................................................... 12

1.4.3 Inhibitors of ACE ......................................................................... 12

1.4.4 Standard Drugs or Synthetic Inhibitors of ACE ........................... 13

1.4.5 Natural Compounds, Peptides or Plant Extracts as ACE

Inhibitors ..................................................................................... 16

1.5 Drug Discovery Process ......................................................................... 24

1.6 Enzyme Kinetics ..................................................................................... 26

1.6.1 Initial Velocity .............................................................................. 26

1.6.2 Michaelis-Menten Kinetics Vmax and Km ...................................... 27

1.6.3 Lineweaver-Burk PIot .................................................................. 28

1.6.4 Dixon Plot .................................................................................... 30

Page 5: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

V

Chapter 2: Mass Spectrometry Techniques for Enzyme Inhibition

Analyses ....................................................................................... 31

2.1 Introduction to Mass Spectrometry ......................................................... 32

2.2 Mass Spectro metry Instrumentation ....................................................... 33

2.2.1 Ion Sources ................................................................................. 33

2.2.2 Mass Analyzers ........................................................................... 40

2.2.3 Detectors ..................................................................................... 43

2.3 Tandem Mass Spectrometry ................................................................... 44

2.3.1 Tandem-in-space Mass Spectrometry......................................... 44

2.3.2 MS/MS by TripIe QuadrupoIes .................................................... 44

2.3.3 MS / MS by Qq TOF Sy stems ........................................................ 46

2.3.4 Tandem-in-time Mass Spectrometry ........................................... 47

2.3.5 Fragmentation and Ion Activation ................................................ 47

2.4 Mass Spectrometry for Enzyme Assay and Inhibitors Screening ........... 48

2.5 Methods of Mass Spectrometry for Enzyme Assay Screening ............... 49

2.5.1 Direct Infusion-Mass Spectrometry (DI-MS) ................................ 51

2.5.2 Ultrafiltration-Ma ss Spectro metry (UF-M S) ................................. 54

2.5.3 Size-Exclusion Chromatography-Mass Spectrometry (SEC-

MS) ............................................................................................. 57

2.5.4 Immobilized Enzyme-Mass Spectrometry (IE-MS) ...................... 59

2.5.5 FrontaI A ffinity Chromato graphy-Ma ss Spectro metry (F AC -

M S) ............................................................................................. 62

2.5.6 Capillary Isoelectric Focusing-Mass Spectrometry (CIEF-

MS) ............................................................................................. 65

2.5.7 Flow Injection Analysis-Mass Spectrometry (FIA-MS) and

High-Performance Liquid Chromatography-Mass

Spectrometry (HPLC-MS) ........................................................... 65

2.5.8 HPLC Based Continuous Flow System-Mass Spectrometry

(CFS-MS) .................................................................................... 68

2.5.9 Matrix Assisted Laser Desorption Ionization-Mass

Spectrometry (MALDI-MS) .......................................................... 69

Page 6: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

VI

Chapter 3: Method Development for Screening of ACE inhibitors

Using HPLC-ESI-QqQ-MS ........................................................... 73

3.1 Introduction ............................................................................................. 74

3.2 ExperimentaI .......................................................................................... 75

3.2.1 Chemicals and Reagents ............................................................ 75

3.2.2 Calibration Curve ........................................................................ 75

3.2.3 Angiotensin Converting Enzyme Assay ....................................... 76

3.2.4 Sample Preparation for LC-ESI-QqQ-MS Analysis ..................... 76

3.2.5 LC-ESI-QqQ-MS AnaIysis ........................................................... 77

3.3 Results and Discussion .......................................................................... 78

3.3.1 Optimization of LC-MS/MS Analysis Conditions .......................... 78

3.3.2 Optimization of Jet Stream Source Using Design Expert ............ 80

3.3.3 Mass Hunter Optimizer for Peptides............................................ 87

3.3.4 Determination of Calibration Curve, LOD and LOQ .................... 89

3.3.5 Enzymatic Reaction..................................................................... 90

3.3.6 Initial Velocity of Enzyme ............................................................ 92

3.3.7 Determination of IC50 of Inhibitors ............................................... 93

3.3.8 Comparison of Captopril and Lisinopril Inhibitory Potential ......... 95

3.3.9 Qualification of ACE Assay ......................................................... 95

3.3.10 Comparison with Reported Methods ......................................... 96

3.4 Conclusion .............................................................................................. 99

Chapter 4: Method Development for Screening of ACE inhibitors

Using MALDI-MS ....................................................................... 100

4.1 Introduction ........................................................................................... 101

4.2 Experimental ......................................................................................... 102

4.2.1 Chemicals and Reagents .......................................................... 102

4.2.2 MALDl Matrix Preparation ......................................................... 102

4.2.3 Calibration Curve ...................................................................... 102

4.2.4 Enzymatic Assay ....................................................................... 103

4.2.5 Sample Preparation................................................................... 103

4.2.6 MALDI-MS Analysis Optimization ............................................. 104

4.2.7 MALDI MS Analysis................................................................... 105

4.3 ResuIts and Discussion ........................................................................ 106

Page 7: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

VII

4.3.1 MALDl Matrix Preparation ......................................................... 106

4.3.2 Linearity Experiment.................................................................. 107

4.3.3 MALDI MS Analysis................................................................... 110

4.3.4 Enzymatic Reaction................................................................... 113

4.3.5 Determination of IC50 of Standard Inhibitors .............................. 115

4.3.6 Comparison of Captopril and Lisinopril Inhibitory Potential ....... 117

4.4 Conclusion ............................................................................................ 118

Chapter 5: Screening of Different Synthetic Drugs, Compounds and

Natural Extracts for Inhibitory Potential Against ACE ........... 119

5.1 Introduction ........................................................................................... 120

5.2 Experimental ......................................................................................... 120

5.2.1 Reagents and Chemicals .......................................................... 120

5.2.2 Enzymatic Reactions for Inhibition Study .................................. 121

5.2.3 Preparation of Inhibitors ............................................................ 121

5.2.4 Sample Preparation and Analysis ............................................. 122

5.3 Results and Discussion ........................................................................ 122

5.3.1 Inhibition AnaIysis of Drugs/Compounds ................................... 122

5.3.2 Inhibition Study of Synthetic Compounds .................................. 150

5.3.3 Inhibition Study of Natural Compounds and Extracts ................ 167

5.4 Conclusion ............................................................................................ 170

References ................................................................................................ 172

Page 8: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

VIII

Acknowledgements

ALLAH (Subḥānahu wa ta'alā) is the one Who is the beneficent, the merciful, we pray to

You and we ask for Your help. All praise is to Him, Who guides me and gives me strength

to pursue and complete this work. Durood-o-Salaam to the most noble and humble

creation of them all, our beloved holy prophet Hazrat MUHAMMAD (Ṣallā llāhu ʿalayhi

wasallama) who enlightened our conscience with the essence of faith in ALLAH (SWT).

I offer my sincere gratitude to the three great scientists: Prof. Dr. Salimuzzaman Siddiqui

(late), Founding Director; Prof. Dr. Atta-ur-Rahman (F.R.S., N.I., H.I., S.I., T.I.), Patron-in-Chief,

International Center for Chemical and Biological Sciences, University of Karachi, and Prof.

Dr. Muhammad Iqbal Choudhary (H.I., S.I., T.I.), Director of the H.E.J. Research Institute of

Chemistry, International Center for Chemical and Biological Sciences, University of

Karachi. Without them there would be no concept of having a world class research

institute in Pakistan. I am especially thankful to the Prof. Dr. Atta-ur-Rahman and Prof. Dr.

M. Iqbal Choudhary, their vision and leadership made possible to transform the H.E.J.

Research Institute of Chemistry into an international center of excellence and made it one

of a kind in world, where I had state-of-art facilities for my Ph.D. work, under single roof.

I cannot write, nor can I explain the invaluable support and encouragement received from

my supervisor cum mentor Prof. Dr. Syed Ghulam Musharraf. His kindness, personality,

knowledge are the reasons which motivated me to pursue the studies of M.Phil./Ph.D. His

attention, supervision and guidance had transformed me from a summer internship

internee to a Doctor of Philosophy (Ph.D.). I can never forget his kind and encouraging

attitude and all of the above his patience, during the whole tenure of my studies and

research, I will remain forever indebted to him.

I am very thankful to Prof. Dr. Khalid M. Khan (S.I., T.I.), for providing compounds for

screening, and Prof. Dr. M. Iqbal Bhanger, Dr. M. Imran Malik and Dr. Sirajuddin for their

valuable time to review my thesis. I am also very thankful to the faculty of the ICCBS. As

an indigenous scholar, I am also thankful to the Higher Education Commission for their

finalcial help.

Simple words are not enough for the two personalities in the world to them I owe

everything, my father Fakhruddin Bhatti and mother Yasmeen Gul. They have supported

me in all ways in my studies, these were their prayers, due to which I was able to achieve

this position in my life. I am also thankful to my big brother and all my family members for

their love, affection, and unparalleled support throughout my life.

Page 9: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

IX

I am very thankful to Dr. Arslan Ali, who always lend his helping hand in solving my

problems. I am grateful to all my seniors Dr. Naghma Hashmi, Dr. Naveed Malik, Dr. Arif

Ahmed, Dr. Aisha Bibi, Dr. Irfan, Dr. Jalal, Dr. Madiha, Dr. Shumaila, Dr. Nayab, Dr.

Shoaib, Ms. Mahwish, Ms. Iffat, Dr. Urooj, Dr. Amna, Dr. Najia and Dr. Qamar ul Arfeen;

my class fellows Ms. Aisha Khalid and Dr. Kashif; and all lab fellows Dr. Faraz, Ms. Aisha

Iqbal, Ms. Fizza, Mr. Hammad, Mr. Saqib, Ms. Wardah, Ms. Ambreen, Ms. Kosur, Ms.

Fareeha, Ms. Sabiha, Ms. Maryam, Ms. Tamkeen, Mrs. Adeeba, Ms. Sindhia, Mrs.

Hamna, Mr. Noman Khan, Mr. Noman, Mr. Ramzan, Mr. M. Zaki, Mr. Saeed, Mrs. Nudrat,

Ms. Naheed, Mrs. Muzna, Mr. Nasir, Ms. Tehreem, Ms. Nida, Mr. Usman, Ms. Zaib and

Mr. Faisal I spent a wonderful and memorable time with them thanks to all of them. Mr.

Shehzad Abbas, our lab attendant deserves special thanks for his help and cooperation.

Last but not least, I am thankful to my dear friends Dr. Ayaz Anwar, Mr. Abdul Mateen,

Mr. Junaid ul Haq, Dr. Itrat Fatima, Dr. Shazia Iqbal, Mr. Saquib Shahbaz and Dr. Zahid

for their unconditional help, care, support and company all the time, and my oldest friend

Mr. Shoaib Khalid who showed me the way towards this achievement.

At last I pray from all mighty ALLAH for brightening future, good health and joyful life to all

my teachers, family members and friends. Aameen

Muhammad Salman Bhatti

21st June 2019, Karachi

Page 10: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

X

Personal Introduction

I was born in Mirpurkhas, “The city of Mangoes” in the south-east of Sindh, I belong to a medium family system in term of finance and education, but my father took a bold step and did his best to support me in getting higher educations. I started my education voyage in 1993 from an Urdu medium Govt. Railway Primary School. Then, I moved to Shah Wali Ullah Govt. High School, and passed my matriculation in 2003. Then I took admission in Shah Abdul Latif Govt. Degree Science College, Mirpurkhas, in pre-medical category and passed in 2005. My aim was to be a medical doctor but in two attempts to medical college admission I left out with a difference of one position, then I took admission in B.S. (Hons.) Chemistry in 2007 and passed in 1st division in Analytical Chemistry from Department of Chemistry, University of Sindh, Jamshoro in 2010. During my final year, I got an internship in H.E.J. Research Institute of Chemistry and worked in the lab of Prof. Dr. Syed Ghulam Musharraf, there I saw another opportunity to fulfill my dream of becoming a doctor, which was, to get a Ph.D. For this purpose, I worked hard and got admission in this prestige institute H.E.J. in June 2011. I did not have a good head start in my early education, as well as I didn’t get high grades or position in my career, but Alḥamdulillāh, with my hard work and dedication I was able to achieve goals of my life. In H.E.J., I joined my respected mentor Prof. Dr. Syed Ghulam Musharraf and started the journey of research under his supervision. My research work was decided on the first day of joining H.E.J., and it was the interface of biological and analytical field, “Development of enzyme inhibition assay on mass spectrometry”. I started to explore different aspects and chose the Angiotensin Converting Enzyme because of its pharmacological importance. I had many down times in my research, and it was very difficult for me to continue the Ph.D., but with patience, hard work and support of my supervisor I developed this work and finally published the developed method in international journal of impact factor 3.5. One of my achievement in H.E.J. is that I obtained 94 percentile in international subjective GRE, which is the second highest percentile, anyone has ever got in this test in H.E.J. During the tenure of H.E.J. I also had the privilege to teach to the students of National Chemistry Talent Contest (NCTC), it was a great teaching experience. I always loved to work on advance instruments or techniques, in this context, I conducted a one-week workshop, under guidance of Prof. Dr. David Smith, entitled “On-hands training on Capillary-HPLC and Nano-flow ESI-MS workshop”, in which we taught how to transform a UPLC into a nano flow-LC and preparation method of capillary columns. Other then scientific life, few of my hobbies are playing table tennis, watching movies, videos, solving puzzles and to surf internet as ‘netizen’. H.E.J. is always like my second home, where I spent wonderful time with friends and collagues and I learned a lot not only about the science but also the social values and ethics of prefossional life, which will help me in various stages of my life. I am serving my country in a federal commission, and I would like to continue serving my country with my knowledge and scientific research in one way or another. I will always cherish this prestigious institute and my supervisor for supporting me at every step and making me what I am today. May ALLAH bless us all Aameen.

Muhammad Salman Bhatti

21st June 2019, Karachi

Page 11: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

XI

List of Publications

1. Musharraf, S. G., Bhatti, M. S., Choudhary, M. I., and Rahman, A.-u. (2017).

Screening of inhibitors of angiotensin-converting enzyme (ACE) employing

high performance liquid chromatography-electrospray ionization triple

quadrupole mass spectrometry (HPLC-ESI-QqQ-MS). European Journal of

Pharmaceutical Sciences, 101, 182-188.

2. Ahmad, I., Bano, R., Musharraf, S. G., Sheraz, M. A., Ahmed, S., Tahir, H.,

ul Arfeen, Q., Bhatti, M. S., Shad, Z., and Hussain, S. F. (2015).

Photodegradation of norfloxacin in aqueous and organic solvents: A kinetic

study. Journal of Photochemistry and Photobiology A: Chemistry, 302, 1-10.

3. Ahmad, I., Bano, R., Musharraf, S. G., Ahmed, S., Sheraz, M. A., ul Arfeen,

Q., Bhatti, M. S., and Shad, Z. (2014). Photodegradation of moxifloxacin in

aqueous and organic solvents: a kinetic study. AAPS PharmSciTech, 15(6),

1588-1597.

4. Bhatti, M. S., and Musharraf, S. G. (2019). Screening of standard drugs

against Angiotensin converting enzyme for their repurposing as ACE

inhibitor. (manuscript in progress)

5. Bhatti, M. S., Musharraf, S. G. and Khan, K. M. (2019). Screening of

derivatives of indole as Angiotensin converting enzyme inhibitors.

(manuscript in progress)

Page 12: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

XII

List of Figures

Figure 1.1: Crystal structure of homosepian testicular ACE with Lisinopril (ESRF; Natesh et al., 2003). ........................................ 4

Figure 1.2: Schematic pathway of RAAS (Mason et al., 2012). .................... 5

Figure 1.3: Renin angiotensin aldosterone system (Wikipedia). ................... 6

Figure 1.4: Risks associated with RAAS (Werner, Pöss, and Böhm, 2010). .......................................................................................... 8

Figure 1.5: Types and mechanism of different enzyme inhibition. ................ 9

Figure 1.6: Graphical illustration and equations for different types of reversible inhibitions (Balbaa and El Ashry, 2012). ................... 10

Figure 1.7: Graph for calculation of initial velocity of enzyme reaction. ....... 26

Figure 1.8: Michaelis-Menten kinetics graph and different types of inhibitions. ................................................................................. 28

Figure 1.9: Lineweaver-Burk plots for different types of inhibition. .............. 29

Figure 1.10: Dixon plots for competitive and noncompetitive inhibition. ........ 30

Figure 2.1: GeneraI schemati c diagram of mass spectro meter (Soderberg, 2019). .................................................................... 33

Figure 2.2: Sche matic diagram of ionization source of MALDl mas s spectrometer (Szot and Croxatto, 2019). .................................. 34

Figure 2.3: Comparasion of Nd:YAG laser and smartbeam spectra (Holle and Ketterlinus, 2008). ................................................... 36

Figure 2.4: Smartbeam setup (Holle et al., 2006). ...................................... 36

Figure 2.5: Process of desoIvation and ionization in ESl-MS (IPFS). ......... 38

Figure 2.6: Jet stream ionization source working principle (Mordehai and Fjeldsted, 2009). ................................................................ 39

Figure 2.7: Thermal profile of jet stream source showing confinement of zoning by super-heated nitrogen (Mordehai and Fjeldsted, 2009). ....................................................................... 39

Figure 2.8: Comparison of spectra using conventional ESI and jet stream technology (Mordehai and Fjeldsted, 2009). ................. 40

Figure 2.9: Transmission of different ions through quadrupole mass analyser (Khan, 2019). .............................................................. 41

Figure 2.10: Ions separation as a function of time in a TOF analyzer. .......... 42

Figure 2.11: Sch ematic of a tripIe quadrupoIe ma ss spectro meter (Jackson, 2019). ....................................................................... 45

Figure 2.12: Different types of mass spectrometry based screening assay (de Boer et al., 2007). ..................................................... 50

Figure 2.13: Different techniques of mass spectrometry used for enzyme assay screening. ......................................................... 51

Page 13: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

XIII

Figure 2.14: Direct Ionization mass spectrometry spectra of enzyme inhibitor complexes (Stokvis et al., 2000). ................................. 52

Figure 2.15: ESl-FT-lCR-MS screening o f library of peptides (J. Gao et al., 1996). .................................................................................. 53

Figure 2.16: Ultrafiltration process in enzymatic assays (de Boer et al., 2007). ........................................................................................ 55

Figure 2.17: α-Glucosidase inhibitory assay with S. baicalensis extract using UF-LC-PDA-ESI/MS (J. Wang et al., 2014). .................... 55

Figure 2.18: UF-MS analysis of COX-II inhibitory assay (Nikolic et al., 2000). ........................................................................................ 56

Figure 2.19: Fast SEC-MS of a library of compounds and identification on MS/MS basis (Annis et al., 2004). ........................................ 58

Figure 2.20: Covalently immobilization of enzyme with the resin surface (Sirisha, Jain, and Jain, 2016). ................................................. 59

Figure 2.21: Enzyme immobilization methods (Sirisha, Jain, and Jain, 2016). ........................................................................................ 60

Figure 2.22: Screening of library of compounds using immobilized enzyme assay, spectra A is before incubation & spectra B is after incubation (H. Gao and Leary, 2003). ........................... 61

Figure 2.23: Principle of FAC-MS where early eluting compounds has no or low affinity and vice versa (Schriemer, 2004). ................. 63

Figure 2.24: FAC-MS enzyme inhibitor screening using ‘roll-up height’ phenomenon (Slon-Usakiewicz et al., 2005). ............................ 64

Figure 2.25: FIA-MS in step A preparation and incubation of sample, in step B injection of sample into LC-ESI-MS system (Luque de Castro, 2019). ...................................................................... 66

Figure 2.26: Spectra and results of inhibition study of drugs using FIA-MS (Takayama et al., 1997). ..................................................... 67

Figure 2.27: Continuous flow system with MS detection using sample injection and online reaction of enzyme and substrate in the presence of inhibitors (Ingkaninan, Hazekamp, et al., 2000). ........................................................................................ 69

Figure 2.28: Multiplex assay on MALDI-MS (A) is the spectra of cοntrοl withοut any inhibitοr, (B) is spectra of with the inhibitοr Lisinοpril (Hsieh, Keshishian, and Muir, 1998). ......................... 72

Figure 3.1: Scheme for samples of calibration curve. ................................. 76

Figure 3.2: Scheme of solvent gradient on HPLC. ...................................... 77

Figure 3.3: Total ion chromatogram of peptides. ......................................... 79

Figure 3.4: Retention time and MRM transition of bradykinin, angiotensin II and angiotensin I, in a standard mixture. ............ 80

Figure 3.5: Response of bradykinin and angiotensin I. ............................... 84

Page 14: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

XIV

Figure 3.6: Comparison of multiple charge ions of angiotensin I (above) and bradykinin (below). ................................................ 86

Figure 3.7: Steps of ‘MassHunter Optimizer for Peptides’. ......................... 88

Figure 3.8: Calibration curve of angiotensin I. ............................................. 89

Figure 3.9: ACE assay with different concentrations of enzyme and time course. .............................................................................. 92

Figure 3.10: Inhibition of ACE by Captopril and Lisinopril. ............................ 94

Figure 4.1: Molecular structure of α-CHCA. .............................................. 106

Figure 4.2: Spectra of α-CHCA on MALDI-MS. ......................................... 107

Figure 4.3: Calibration curve for %conversion. ......................................... 110

Figure 4.4: Parameters of analysis method. (A) sample carrier, (B) detection parameters, (C) Spectrophotometer settings and (D) instrument digitizer and detector settings. ......................... 112

Figure 4.5: MALDI-MS peaks detection and processing parameters. ....... 113

Figure 4.6: Graph for ACE reaction without any inhibitor. ......................... 115

Figure 4.7: Scheme for analysis of inhibitor samples and control in well plate. ....................................................................................... 116

Figure 4.8: Inhibition of ACE by Captopril and Lisinopril. .......................... 117

Figure 4.9: Comparative MALDl-MS spectra of different concentration of Captopril and Lisinopril. ...................................................... 118

Page 15: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

XV

List of Tables

Table 1.1: Standard drugs or inhibitors of ACE. ............................................. 14

Table 1.2: Different plant extracts as active inhibitors against ACE. .............. 17

Table 1.3: Peptides from different food proteins as ACE inhibitors. ............... 17

Table 1.4: Flavonoids and their derivates from natural sources, as active ACE inhibitors. ............................................................................... 19

Table 3.1: MRM transitions and source conditions for fragmentation. ............ 78

Table 3.2: Range of different parameters feed to the software. ...................... 81

Table 3.3: Experiments design output of Design-Expert................................. 82

Table 3.4: Response of bradykinin and angiotensin I correspond to each experiment. .................................................................................... 83

Table 3.5: Checking of ionization of multiple charge ions of angiotensin I and bradykinin. .............................................................................. 85

Table 3.6: Intraday and Interday accuracy and precision determination of angiotensin I. ................................................................................. 90

Table 3.7: Initial velocity of ACE in enzymatic assay. ..................................... 93

Table 3.8: Intraday and interday precision and reproducibility of ACE inhibition by Captopril and Lisinopril. ............................................. 96

Table 3.9: Comparison of the current developed method with previously reported methods. ......................................................................... 97

Table 4.1: Different spotting methods on MTP. ............................................ 104

Table 4.2: Sample scheme of linearity experiment. ...................................... 108

Table 4.3: Percentage conversion of different samples for linearity experiment. .................................................................................. 109

Table 4.4: ACE reaction without any inhibitor. .............................................. 114

Table 5.1: All drugs or cοmpounds used for the inhibitions istudy. ............... 123

Table 5.2: Screening results of drugs/compounds, IC50 values of active drugs. .......................................................................................... 147

Table 5.3: Ten most active drugs along with their purposes. ........................ 150

Table 5.4: Structures of base compounds and general structures of derivatives classes. ..................................................................... 151

Table 5.5: Synthetic compounds used for inhibition study. ........................... 153

Table 5.6: Screening results of synthetic compounds, IC50 of active compounds. ................................................................................. 166

Table 5.7: Natural compounds used for inhibition study. .............................. 168

Table 5.8: Natural extracts of plants used for inhibition study. ..................... 169

Table 5.9: Results of natural compounds and extracts, IC50 of active samples. ...................................................................................... 170

Page 16: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

XVI

List of Abbreviations

Terms Abbreviation

%C Percentage conversion

%IA Percentage inhibitory activity

%MA Percentage maximal activity

[I] Concentration of inhibitor

[S] Concentration of substrate

ACE Angiotensin Converting enzyme

ADH Antidiuretic hormone

AI / AngI Angiotensin 1

AII / AngII Angiotensin 2

α-CHCA / HCCA α-Cyanο-4-hydrοxycinnamic acid

BK Bradykinin

DMSO Dimethyl sulfoxide

DTT Dithiothreitol

EIC Extraction-ion chromatogram

ESI Electrospray ionization

FAC Frontal affinity chromatography

FIA Flow injection analysis

HPLC High performance liquid chromatography

IC50 Inhibitor concentration used to reduce enzyme activity up to

50%

Km Concentration of substrate at half of maximum velocity

LOD Limit of detection

LOQ Limit of quantitation

m/z Mass-to-charge ratio

Page 17: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

XVII

Terms Abbreviation

MALDI Matrix-assisted laser desorption ionization

MI Myocardial infection

MRM Multiple reaction monitoring

MS/MS / MS2 Tandem mass spectrometry

MTP MALDI target plate

mU Milli Units (concentration of enzyme)

QqQ Triple quadrupole

RAAS Renin angiotensin aldosterone system

RSD Relative standard deviation

SEC Size exclusion chromatography

TIC Total ion current

ToF Time-of-flight

UF Ultra filtration

Vmax Maximum initial velocity of enzyme

vo Initial velocity of enzyme

Page 18: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

XVIII

Summary

Enzymes play a very crucial role in the regulation of almost all process of life. Enzymes

are also an important class of drug target; screening of inhibitors for enzymes is the first

step in drug discovery process. Angiotensin converting enzyme (ACE) is a

pharmacological important enzyme, it converts the Al into All. It plays a key role in Renin

angiotensin aldosterone system (RAAS), which regulate the blood pressure and

hypertension. Over expression of RAAS can cause many cardiovascular and renal

diseases, therefore, inhibition of ACE is a promising way of controlling over expression of

RAAS.

Many methods have been used for the inhibition study of ACE including

spectrophotometric, fluorimetric, HPLC, mass spectrometric, etc. When LC-MS combines

with the MS/MS then it becomes very unambiguous and sensitive analysis techniques for

detection and identification of peptides in complex samples. In this study, a new sensitive

and robust HPLC-ESI-MS/MS method, with very low limit of quantitation was developed

for ACE screening assay. In this method, substrate was used instead of product to

determine ACE activity. For this purpose, a calibration curve of Angiotensin I was

developed in a range of 20-200 nM, linear equation y=0.0888x-0.1932 and R2=0.999. Two

commercially available antihypertensive drugs, Captopril and Lisinopril, were checked to

validate the method and their IC50 values were found to be 3.969 μM and 0.852 μM,

respectively. This newly developed method required very low amount of substrate,

enzyme and inhibitor per sample.

MALDI was also explored for the potential of enzyme inhibition study, MALDI-TOF-MS is

a well-known technique for the analysis of protein and peptides. Relative quantification

was performed by using the ratio of substrate and product. In this study, a high-throughput

method of ACE inhibition was developed on MADLI-TOF-MS. In this method, a linear

curve of AI was developed for 0-100% conversion, linear equation y=9.3175x+3.4177 and

R2=0.994. Both standard inhibitors, Captopril and Lisinopril, were checked for their

inhibition and their IC50 values were found to be 8.256 μM and 1.962 μM, respectively,

which validated the method. This method provides a fast and alternative for the screening

of drugs to find their potential against the target enzyme.

As a final step, 77 commercial drugs, 40 synthetic compounds, 4 natural products and 3

extracts of plants were investigated for their inhibitory potential. In drugs, 36 showed

moderate to good inhibition of as low as IC50=272 µM. In synthetic compounds, 32

compounds showed significant inhibition ranged from IC50 = 320-5123 µM. In the last

category, all 4 compounds were moderately active with IC50 value 496-9705 µM; while

only one plant extracts showed some inhibition with IC50 =10779 µg/mL (ppm).

Current study has proved that direct nature of the MS measurement ensures the

minimization of the false results and large number of compounds can be screened without

the need for labels or additional internal standards, both MALDl-TOF-MS and LC-ESI-

MS/MS are valid readout for enzyme inhibitor screening assays.

Page 19: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

XIX

خلاصہ

�� �� � �� �م � ���ف �� ��ر �� � ا�� �� ا��� �� !"

#� $% &' �ت �-,ے ادو�� /۔ %" �� �� � �� دار ادا � 01 ا�� $2 / 5�ل ��3 7�م 6" �8 � $� �:, �; �< �� &=� �-,ے ز�" "% ?@ ": AB � $C /

�۔ �D م� �E FG �2 � H7I �� شF �� &' �ت �، Angiotensin Converting Enzyme (ACE) ادو�� �D ہ,-� "% � � ا�� � ا�� A^_�`a &�b� ادو��

� $CAI �dAII A�e ۔� �D � �� H ���$� �: / ��3RAAS �d �ن "C ر� ij "k � "�� �واور �$ �ن �� د�$ "C � $C � �D � �� دار ادا � � ا�� / ا�� �۔ ��3 �D � �� �

"l "mnRAAS ��

�۔ �D � �� "o $p q$rs � 7�v�ر��uں $: &' �م " "< �wj "x3 �y دے اور انz �، دل اور "�{|$� � G�ر �، G�ACE~ا ا{" �d رو �" H7I ��RAAS , ��1 $2 � � ا�� � "�� �� �$u� �E ��

�۔ �D Aرا��� �

ACE ع� "� �o3ا ���y 01 $2 � ��� �� ��� � ���7�ل �msا � ��� < ,� ��5 �mn �y &

b �7�v �: ��m۔�/ �� �� q $�LC-MS �dMS/MS ��� $% � �� �wj "x3 �y ��u �

0 "�� "� i� اور � "���_ A�� ���س �� :� ���o �� �ہ :� �� �� �� �: A�e � � �j�س �� /، ا�� ��3 A^_�`a ۔ اس� �D � �� � $% �$� ��� "o� �� &' A�e� $ �

# �¡$vn , �� j� 01�س اور ¢" $2 ��HPLC-

ESI-MS/MS ¥ �¦�§ �� 0�r¨F© &' � "� �ار ��5aم �ªa � �� �% �« 01 �ر، $2 A �۔ �,>�¬� ��� � ��� "� $: / ��3 A �¬ � '& ®,z­ACE ?اس �,>� ��� �� � ��¯�< ��

��� ا§°& $± �²�ت �� #$ "³´a ۔� ��� � �7�ل ���msا �d �-,ہ �� ز��%"� �� �µ �¶a اسAI ۔� ��� � �ر �� �� �: ? $@

"�·,¸ �6 � �ری � ا�� ��ºa دو ��� �ل �� ��|�� !"

#� $% &' � ��� < اس �,

�ت °& Lisinoprilاور Captoprilادو�� / ا�" ��3 ¾ $� � ��� � �7�ل ���msا �d50IC q�r �:, ��Á� �ار �$ �Eا &'Mμ3.969 اورMμ0.852 &' ��5aم

�?۔ @� / ��3 A �¬ دہ �,>� �ر �� �: � �� ": �-,ہاس �-,ہ، %" �۔ ز��%" ��� � �7�ل ���msا � �ار �ªa �« 01 $2 &' �ع "� �o3اور ا

� ��� �� �È�`a ?É� "� �o3ا &' �-,وں "%MALDI ۔� ��� � �7�ل ���msا ?Ë $¦ �dMALDI-TOF-MS ت اور� ��ÏÐÑ وف,Òa � � ا�� ��� �� A�e� $ 

��ز �� # ��� ���o �� �:

�ار �ªa &Óذ�� / ��3 A^_�`a ۔اس� �D ��� "o� �� ��� �� � "� �²�ت اور��5aم "³´a � ��� � �ر �� �� �: A �¬�<,� � �� �Õ اور &Ó²ا � ��� ا�� �Ö � �� �7�ل �msا � q�� "� �-,ہ �� :� ز��%"

،/ ��3 A �¬ ×�۔ اس �,>� �¦AI اور � ��� � �ر �� �� �: ? $@"�·,¸ �6 � � ا�� &' ��³ اس

i:��u ت� �ری ادو�� ��ºa دو ? �Øو ��� �� Captopril اورLisinopril � �7�ل ���msا �d

&° / ا�" ��3 ¾ $� � ���50IC q�r �:, ��Á� �ار �$ �Eا &'Mμ8.256 اورMμ1.962 ۔?� @� &' ��5a �dم �ر A �¬ �تاس �,>� �? ادو�� @ �ر اور :" �� "Úر � �� �Õ � � ا�� ��� �� !"

#� $% &'

�۔ �D � ��Û§� $% � �7�ل ���msا ر ���� �� A �$�دل �,>�¬� �o3 �

�م �� ��ر ��، �E ?Ü� v¨77 ب اد� �� �osدq ,- &�b� �²�ت اور ��uدوں �� q ,- ?É�"³´a4، 40، و�� "³´a &

�bر� �E3 à �� ��³ ,ق

�âã �# ��� �� 0�r¨F© 5�ل "6 , �� "¢ &' ان

�y / ��3 ¾ �@?۔ �$ �ب اد 36'& �� �osد q ,- &�b� »� و�� �y �« / ��3 ¾ $� ،&' , �ä� "å ?­z,® ال� ��æا � "� 50IC ار� �Eا &' Mμ= 272 50IC ،?@� &

b� ��

�y / �É? -, �$�ت ��3 "³´a32 / ��3 � $è &' , �ä� "å ?­z,® ں� ���7 "8 ��ار 50IC-, �$�ت �" �Eا &'Mμ320 �yMμ5123 ۔? �Øر � �� ،/ ��3 �é

�k ی "êآ

7�م � -, �$�ت �84 "� 50IC ر� �E &'Mμ496 �yMμ9705 a ¥ �¦�§ �� � � ��uدے �� à,ق �" �$ì Aí,ف ا�� $ '&۔ , �ä� "å یz,® ل� ��º= 50IC

10779 μg/mL (ppm) .&' , �ä� "å ?­z,® ¥ �Ðî ¥ �¦�§ �� �

Ó& دا%"

"ï� � ا©" �� ðñ$� �oò اور � �D &

�b� "� $: ?" @ �o �ó�< �d ?Ü« &' ô

�#� �� ": õö ¸,ت ²" "6 ? i÷

�ø�7�v '& �$اہ را�0 :� ¾�øا � �ùا AB � �D ا� �Ö 0 $:� i� �y A^_�`a H7úû اس &' �روں ��ºa &

&' � �� -, �$�ت "��7�v , �$|ے :� �� "ü$< �� ورت, "ì!

"#� �۔ %$ �D ?� @Û§ � $% &'MALDI-TOF-MS اورLC-ESI-MS/MS � ��� � ���ر �� دوu"�ں �� :�

،A �¬�<,� 0�ýþ اور Hزوں ، ا���� H�ö $� ��� �� A^_�`a ?É� "� �o3وں �� ا,-� /۔%" �� �� � ��� < ,� �Èوا � "� �: �$A د ���o "p

Page 20: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

1

Chapter 1: Pharmacological Importance of

Enzymes and Angiotensin Converting Enzyme

(ACE)

Page 21: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

2

1.1 Enzymes in Life: Brief History

Till the start of 19th century, it was considered about the processes like milk

souring, formation of alcohol from fermentation of sugar could only be happen

because of some Iiving organisms. ln first quarter of 19th century, the key

substance ‘diastase’ was isolated which was responsible for breaking the sugar

and it is also known as amylase. After a while, another substance was extracted

from the gastric juice and termed as ‘pepsin’, it was responsible for the digestion

of dietary proteins.

These types of many other substances were given the name of ferments. It was

suggested by Justus Von Liebig, that, these ferments may be of nonliving

materials which were obtained from the living cells, but some scientists like Louis

Pasteur and others still uphold the concept the ferments that these feremnet must

have some living materials inside them. Gradually, the term ‘ferment’ was replaced

by the ‘enzyme’. Wilhelm Kfthne proposed this term of enzyme 1st time in 1878, it

was originated from the word of Greek ‘enzumé’, which means “in yeast”. It was

the factor from the yeasts that was revealed and clear the dispute in favor of

nonliving theory of catalysis. In 1897, It was shown by Hans Buchner and Eduard

Buchner that, extract of yeast cell can also cause the fermentation of sugar even

though no living cells were present in it (Palmer and Bonner, 2007).

Enzymes play a very important role in the living beings by regulating processes as

biological catalyst. The use of enzyme is not only, limited to the biological process

but it is widely used in industrial processes as well as enzymes are using as target

in drug discovery process. The importance of enzymes from natural and synthetic

source is increasing rapidly (Liesener and Karst, 2005).

1.2 Enzyme Catalysis

Enzymes are biological catalysts which increases the rate of reactions which are

taking place in living cells by providing alternate pathways, without getting any

chemical change to themselves. The reactant of an enzyme catalysed reaction is

called as “substrate” and it is converted into the “product” by specific reaction.

Every enzyme is very specific in its function, utilize its specific substrate and in

Page 22: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

3

result produce its specific product. Enzymes are specific type of protein, but

sometimes, specific types of enzymes need one or more non-protein component/s

to perform their function, this non-protein component is known as coenzyme or

cofactor. A coenzyme can be an organic molecule or be a metal ion (Palmer and

Bonner, 2007).

Enzymatic reactions play a very important part in the regulation of nearly all the

processes of living organisms, but applications of enzyme is not limited to that,

enzymes now a days are used in industrial processes as biocatalysts or can be

used as targets to screening the drugs, enzymes from both synthetic and natural

sources are getting more important day by day. (Liesener and Karst, 2005).

1.3 Αngiotensin Converting Εnzyme (ΑCΕ)

Αngiotensin converting enzyme (ACE) is a dipeptidyl carboxypeptidase (E.C.

3.4.15.1) which has a zinc (Zn) atom and having 150-180 kDa range of mass. ACE

is comprised of one polypeptide chain which have two terminal domains of N and

C. In each domain there are two sites for catalysis reaction. ACE is a

metallopeptidase which produce the Angiotensin II from inactive Angiotensin I by

cleaving its two peptides histidine–leucine from the C-terminal, crystal structure of

human ACE is shown in Figure 1.1 (Balasuriya and Rupasinghe, 2011; Lapointe

and Rouleau, 2002; Ortiz-Salmerón, Barón, and Garcı a-Fuentes, 1998; Shi et al.,

2010).

ACE is usually present on the outer surface membrane of a cell so that, active site

of ACE is accessible by the foreign environment of cell. ACE shows its expression

in various tissues and organs; the heart, the brain tissues and its several regions,

renal proximal tubular endothelium, vascular endothelium, the lungs, activated

macrophages are included in them (Lapointe and Rouleau, 2002). ACE present in

the lungs has the highest concentrations. Other than lungs ACE is also present in

vascular endothelium, renal proximal tubules, gastrointestinal tract, and the

tissues of heart and brain, in low concentration. It exists in both ways as a

circulatory (globular) enzyme and membrane bound enzyme (Alves et al., 2005;

Balasuriya and Rupasinghe, 2011).

Page 23: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

4

Figure 1.1: Crystal structure of homosepian testicular ACE with Lisinopril (ESRF; Natesh et al., 2003).

ACE mainly perform transformation of AI into its product AII; by cleaving the C-

terminal dipeptide (L-histidyl-L-Leucine) of AI, generating the physiologically active

vasoconstrictor peptide AII. On the other hand ACE also act of the Bradykinin and

degrade it by cleaving at 7-8 peptide bond, bradykinin is a potent vasodilator

(Elased et al., 2006; Menard and Patchett, 2001). Different studies have shown

that the ACE has more Km (affinity to react with substrate) with AI in comparasion

to the bradykinin (BK) (Lapointe and Rouleau, 2002; Wong, 2016). ACE is a core

component of Renin Angiotensin Aldosterone System (RAAS), maintaining the

body’s blood pressure and its regulation is the main function of this system

(Jianping Wu, Aluko, and Muir, 2002).

1.3.1 Renin Angiotensin Aldosterone System (RAAS)

Renin Angiotensin Aldosterone System (RAAS) is an essential body fIuid

maintenance system which has the basic function of stabilizing the blood pressure,

it is also called as Renin Angiotensin System (RAS). It was discovered over a

Page 24: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

5

century ago, it is a biochemical pathway which performs very critical functions in

physiology of cardiovascular system, it is also responsible in various steps of

pathways in diseases related to cardiovascular and cardio-renal (Elased et al.,

2006; Lu, Liu, and Feng, 2011).

The pathway of RAAS starts with the production of angiotensinogen, it is an α-2

globulin which is produced from the liver, it is a sequence of 452 aminoacids but

only the 10 first amino acids plays significant part in RAAS, while angiotensinogen

does not have any independent function by itself. Next important protein in RAAS

is Renin, it is produced from ‘prorenin’ which is an enzymatic precursor and

secreted from the kidney juxtaglomerular apparatus into the bloodstream. Once

the renin is activated it perfrom action of angiotensinogen and cleaves its 10

peptides chain Asp-Arg-VaI-Tyr-IIe-His-Pro-Phe-His-Leu to produce the

Angiotensin I. Like angiotensinogen, AI is also not active, but it acts as the

substrate of Angiotensin converting enzyme (ACE). ACE cleaves two residues,

Histidyl and Leucine, from AI to produce octapeptide Angiotensin-ll (AII) (Asp-Arg-

VaI-Tyr-IIe-His-Pro-Phe), schematic pathway of RAAS is shown below in Figure

1.2 (Mason et al., 2012).

Figure 1.2: Schematic pathway of RAAS (Mason et al., 2012).

AII is the main peptide component of the RAAS, it acts different ways as intracrine

hormone, paracrine/autocrine hormone and endocrine hormone. AII, which is

converted from AI by ACE, is the main component which have effect in RAAS. Its

biological actions are produced through the selective binding of this peptide to

Page 25: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

6

different types of receptors, it causes the vasoconstriction (constriction of blood

vessels) by interacting with receptors of angiotensin on cells of vascular smooth

muscles, it also stimulate the adrenal cortex of kidney to release the aldosterone

which increases the water retention in blood, another function of the AII is to

stimulate the pituitary gland to secrete the ADH which also increase the water

reabsorption, these all factors ultimately leads to the high blood pressure. Effects

of RAAS are illustrated below in Figure 1.3 (Elased et al., 2006; Lapointe and

Rouleau, 2002).

Figure 1.3: Renin angiotensin aldosterone system (Wikipedia).

1.3.2 Problems Associated with Over Expression of ACE and

RAAS

Physiological imbalances of the RAAS may occur within the system or it may occur

by inherited or acquired mutation which can leads to the disturbance in the function

of various components. Mechanical stress to the body can also cause the changes

in related physiological systems which then can disturb the balance of RAAS

(Mason et al., 2012), different genetic factors can also cause the increase activity

of the RAAS and also the sympathetic nervous system, effect of salt on blood

pressure (salt sensitivity) can also effect RAAS. Other factors like environmental

factors, obesity, excess intake of salt in diet and sedentary (deskbound) type of

lifestyle have also impact on disbalance of RAAS (Carey, 2015). The latest Heart

Page 26: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

7

disease and stroke statistics of 2017 by American Heart Association (AHA) were

shown that, in USA, the rate of heart failure (HF) has been increased to 0.8 million

new cases in last 5 years, It was also speculated in the report, that, the number of

patients with HF is expected to rise by 46% by 2030 (MEMBERS et al., 2017).

Traditionally, pathological upregulation of RAAS are primarily associated with

cardiovascular risk because of the increase activity of A-II.

Hypertension is the world’s most ubiquitous progressive disorder due to over

activation of RAAS; which can lead up to several other chronic diseases such as

diabetes, stroke, renal problems and most commonly cardiovascular disease. 25%

of total adult population of the world is effected by the hypertension, and it may

increase up to 29% by 2025 (Balasuriya and Rupasinghe, 2011). In many

communities of the world approximately one adult out of three have hypertension,

which is leading risk factor for disability and death (Carey, 2015).

It is been found out that RAAS is involved in progress of different conditions, from

diseases related to musculoskeletal to Alzheimer and various types of cancers

(Mason et al., 2012). In heart related problems most of the cardiovascular

diseases are related to the increased activity of ACE (Lu, Liu, and Feng, 2011).

More exposure to the cardio vascular risk factors can leads to the atherosclerosis,

left ventricular hypertrophy (LVH) or microalbuminuria. Ultimately which can lead

up to the more severe syndromes such as, myocardial infarction (MI), heart stroke

or kidney dysfunction, progressive towards damage of organs and finally leading

to heart failure, final stage of renal diseases, dementia or even death. In

industrialized developed countries these diseases are most common cause of

morbidity and death (Werner, Pöss, and Böhm, 2010). It was also reported by

Lapointe that if ACE has an increase activity in plasma then, the person has an

increased risk of myocardial infarction (MI) and various other cardiovascular

related diseases, like dilated cardiomyopathies and hypertrophic etc. (Lapointe

and Rouleau, 2002).

Page 27: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

8

Figure 1.4: Risks associated with RAAS (Werner, Pöss, and Böhm, 2010).

Above Figure 1.4: Risks associated with RAAS (Werner, Pöss, and Böhm,

2010).Figure 1.4 showed the diseases progression in renovascular system in

lower curve and cardiovascular system in upper curve, due to over expression of

RAAS. Cardiovascular risk factors like blood pressure, diabetes, dyslipidemia, and

smoking etc. facilitate the progression of atherosclerosis, which leads to the

disease of coronary arteries, ischemia and acute coronary thrombosis, to acute

coronary syndromes and myocardial infarction (MI). Damage to the cardiac

structure over the time, decrease its contractility which then lead to the dilation of

left ventricular, which then cause the chronic heart failure and, ultimately cause

the death in early ages. Dysfunction of renal endothelial may be followed by

microalbuminuria (minor increase in the urine albumin level), macroproteinuria

(albumin level more than its upper limit), nephrotic syndrome and, finally, end-

stage renal disease (Werner, Pöss, and Böhm, 2010).

1.4 Enzyme Inhibition

In the body, most of the diseases or at least their symptoms, usually came from

the excess or deficiency of specific metabolite, it may be an influx of foreign

organism, or abnormal growth of cells. If that disturbed metabolite can be

normalized, then these diseases may be cured, and these foreign organisms or

abnormal cells may be destroyed. Most of these diseases and problems can be

countered by inhibiting the activity of specific enzymes (Silverman and Holladay,

2014).

Page 28: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

9

Any factor or compound that can reduce the activity of an enzyme by blocking their

chemical action is called as an enzyme inhibitor. Many drugs or compounds are

enzyme inhibitors because they correct or reduce the metabolic imbalance of an

enzyme by blocking their activity. Enzyme inhibitor are also referred as enzyme

inactivator, it is a common misconception that all those compounds which can

binds with the enzymes are inhibitors; on the contrary, enzyme activators are those

compounds which binds with the enzymes and increase or facilitate their

metabolism (Balbaa and El Ashry, 2012; Silverman and Holladay, 2014).

With respect to the binding of inhibitors with enzymes, there are two main types of

inhibition; reversibIe and irreversibIe inhibition. Furthermore, the reversibIe

inhibition is classified into different types as competitive, noncompetitive and

uncompetitive or mixed inhibition, as shown in Figure 1.5.

Figure 1.5: Types and mechanism of different enzyme inhibition.

1.4.1 Reversible Inhibition

In reversible inhibition, inhibitors bind with the enzyme non-covalently (through

interaction). In this inhibition, inhibitor may change the shape of enzyme but do no

change them chemically. Inhibitors can be bind with the only enzyme, the complex

formed by enzyme and substrate, or both. This attachment of inhibitor leads

toward the various types of reversible inhibition which includes competitive,

noncompetitive and uncompetitive inhibition, sometimes a mixed type of inhibition

may also be formed. The basic pattern of inhibition can be studied by Lineweaver-

Page 29: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

10

Burk. & Dixon plots and Michaelis-Menten equation. A graphcical illustration of

different types of reversible inhibition is shown in Figure 1.6 (Balbaa and El Ashry,

2012; Silverman and Holladay, 2014).

Figure 1.6: Graphical illustration and equations for different types of reversible inhibitions (Balbaa and El Ashry, 2012).

1.4.1.1 Competitive Inhibition

It is the most common enzyme inhibition, in this inhibition both substrate and

inhibitors compete to attach on the active site of enzyme. Competitive inhibitors

usually have similarity in structure with the substrate, therefore they can easily

bind to the substrate attaching site (active site) and blocked the substrate.

Generally, these inhibitors rapidly attain binding equilibria the target enzyme,

therefore inhibition can be observed in the start monitoring of inhibition assay.

Competitive inhibition can be reduced by using a high concentration of substrate,

because the inhibitor and substrate both cannot bind to the active site at a time,

only one of them can bind with the enzyme, therefore high concentration of

substrate is in the favor of enzyme activity.

Page 30: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

11

1.4.1.2 Non-Competitive Inhibition

In this inhibition, inhibitor compounds bind with the enzyme to decrease its activity

but did not affect the site of substrate binding. Inhibitors interact with the enzyme

other than the site of substrate binding (allosteric binding site) which results in the

inhibition of enzyme reaction. When an inhibitor binds at the allosteric site, in

results it often produce a conformational change in the structure of enzyme, this

conformational change creates steric hindrance to substrate to binding with the

active site, therefore substrate cannot bind properly to the enzyme.

1.4.1.3 Uncompetitive Inhibition

In uncompetitive inhibition, inhibitor only binds with the enzyme-substrate complex

(E-S complex) it is also known as anti-competitive inhibition. When an enzyme

combines with its substrate, it may open up a site which was hindered before the

complex formation, inhibitors binds at that open site and formed an enzyme-

substrate-inhibitor complex (E-I-S complex), in result that enzyme cannot perform

its function which results in its inhibition. Uncompetitive inhibition usually observed

in those reactions, where 2 or more substrates or products are involved in reaction.

1.4.1.4 Mixed Inhibition

Mixed inhibition is a complex form of inhibition which is not usually observe in

enzyme kinetics. In this case inhibitor can bind with whether of the two states of

the enzyme, when enzyme is in free state and not bind with the substrate or when

enzyme has formed E-S complex, but in both states, inhibitors may have higher

affinity for either one of them. It is called as “mixed inhibition” because, theoretical

it is a mixture of both competitive and uncompetitive inhibition. If the inhibitor has

same ability to bind with the enzyme in both cases regardless of that, enzyme is

free of formed a complex, then it is called as a non-competitive inhibitor.

Sometimes, noncompetitive inhibition is considered as a distinct case of mixed

inhibition.

Page 31: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

12

1.4.2 Irreversible Inhibition

If an inhibitor binds with the enzyme irreversibly (i.e. usually covalently) then, that

inhibition is knows as irreversible inhibition. These inhibitors usually form a

covalent bond with active side of enzyme and generally change it chemically,

however, few specific inhibitors which have tight and slow binding with enzyme

usually do not form a covalent bond with the enzyme but functionally they are

classified as irreversible inhibitor because of their tight binding. In this inhibition,

maintaining a specific concentration of inhibitor is not necessary to sustain the

enzyme-inhibitor complex, because inhibitors perform irreversible reaction (i.e.

covalently) once the inhibitor bind with the enzyme and formed the E-S complex

then it cannot dissociate (there are few exceptions). Once the complex is formed

then enzyme will remain inactive even if there is no additional inhibitor is added

(Silverman and Holladay, 2014).

1.4.3 Inhibitors of ACE

ACE plays a significant role in RAAS therefore, inhibition of ACE become a main

target to achieve the control on hypertension and its related disease and it seems

to be beneficial to fight the various cardiovascular and renal diseases (Balasuriya

and Rupasinghe, 2011; Lapointe and Rouleau, 2002). Research on finding

potential inhibitors of ACE has become expanded broadly and includes the various

classes of compounds from both synthetic and natural product derivatives.

Synthetic ACE inhibitors are the first choice and most effective therapeutics drugs

since decades, marketed available drugs such as, Lisinopril, Captopril, Rampiril

and Enalpiril, are examples of drugs used as ACE inhibitors. Research has also

been carried out to quantify the important ACE inhibitors like Quinapril, Moexipril

in human plasma (Balasuriya and Rupasinghe, 2011; Karra et al., 2012; Lu, Liu,

and Feng, 2011). However, the usage of these drugs for a long period of time can

cause antagonistic side effects like angioneurotic edema, coughing, dizziness etc.

therefore, research to explore the news drugs as a replacement of available drugs

is needed to be continue (Chen et al., 2013; Lu, Liu, and Feng, 2011).

Different classes of natural compounds, food derivatives, plant depravities and

others have been explored for their inhibitory potential against ACE. Derivatives

Page 32: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

13

of food protein which are an important class of compounds, are explored and found

to have potential for inhibition of ACE. Food proteins derivatives can be classified

ainto 3 groups plant derived, animal derived, and microorganism derived.

Category of animal derived comprises of casein, whey protein, ovokinin which

were reported to be effective ACE inhibitors. Proteins from meat and fish were

hydrolyzed by various enzymes like chymases, etc. and resulting fractions were

screened against ACE and found to be active. Plant derived peptides from rice,

flaxseed, soybean, corn and sunflower were also founds to be active inhibitors of

ACE.

Some polyphenolic and terpenoids compounds including flavanols, flavonoids,

flavovols, isoflavones, anthocyanins, hydrolysable tannins, triterpenes etc. were

also reported to be active as natural inhibitors of ACE. Many studies have also

reported that those plants which have high phytochemicals in extracts, were also

the effective inhibitors of ACE (Balasuriya and Rupasinghe, 2011; Hsieh,

Keshishian, and Muir, 1998).

In natural sources, first important source were snake venom peptides (Menard and

Patchett, 2001). In 1965, It was reported by Ferreira (S. Ferreira, 1965) that an

extracted mixture of peptides from the venom of Bothrops jararaca snake (a pit

viper specie in South America) has the potential to stop the action of bradykininase

on bradykinin. Later, Bakhle et.al. (Bakhle, 1968; Bakhle, Reynard, and Vane,

1969) demonstrate that these peptides can also be used to inhibit the activity of

ACE to strop or reduce the production of AII from AI. After that, 9 different peptides

were isolated from this venom which were active against both bradykiniase and

ACE; among them a pentapeptide (Pro-Ala-Trp-Lys-Pyr, where Pyr is l-pyro-

glutamate) was recognized (S. H. Ferreira, Bartelt, and Greene, 1970; Silverman

and Holladay, 2014).

1.4.4 Standard Drugs or Synthetic Inhibitors of ACE

Inhibitors of ACE are classified into 3 groups on the basis of their molecular

structure, these are Dicarboxylate-containing, Sulfhydryl-containing and

Phosphonate-containing group. Dicarboxylate-containing group is the largest

group of ACE inhibitory drugs and it includes Benazepril (Lotensin), Cilazapril

Page 33: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

14

(Inhibace), Enalapril (Renitec / Vasotec), Imidapril (Tanatril), Lisinopril (Listril /

Lopril / Novatec / Prinivil / Zestril), Perindopril (Aceon / Coversyl), Quinapril

(Accupril), Ramipril (Altace / Ramace / Ramiwin / Tritace) and Trandolapril

(Gopten / Mavik / Odrik). Other 2 groups are smaller, Sulfhydryl-containing

inhibitors group have 2 inhibitors Captopril (Capoten) and Zofenopril (Zofecard);

and Phosphonate-containing inhibitors group has only one inhibitor Fosinopril

(Monopril).

Table 1.1: Standard drugs or inhibitors of ACE.

S.No. Inhibitor Molecular weight

Structure

1 Captopril 217.283

2 Zofenopril 429.549

3 Enalapril 376.453

4 Ramipril 416.518

Page 34: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

15

5 Quinapril 438.524

6 Perindopril 368.474

7 Lisinopril 405.495

8 Benazepril 424.497

9 Imidapril 405.451

Page 35: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

16

10 Zofenopril 429.549

11 Trandolapril 430.545

12 Fosinopril 563.672

1.4.5 Natural Compounds, Peptides or Plant Extracts as ACE

Inhibitors

A variety of different classes of natural compounds, plant extracts and peptides

have already been screened for their potential as ACE inhibitors, few classes are

mentioned in previous section ‘1.4.3 Inhibitors of ACE’, which includes but not

limited to flavonoids, peptides, flavanols, etc. Few compounds and peptides from

natural origin, and plant extracts which were found to be active against ACE are

given below in Table 1.2, Table 1.3 and Table 1.4. IC50 values of these

compounds are not giving in this text because different compounds were screened

with different methods, which cannot give a good comparison of their potential.

Page 36: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

17

Table 1.2: Different plant extracts as active inhibitors against ACE.

S.No. Plant Extracts

1 Hibiscus sabdariffa (Hibiscus)

2 Camelia synensis (green tea)

3 Vaccinium ashei reade (Blueberry leaf extract)

4 Vaccinium myrtillus (Bilberry)

5 Senecio inaequidens (A perennial herb)

6 S. ambiguous subsp. Ambigus (ethyl acetate extract)

7 S. ambiguous subsp. Ambigus (n-hexane extract)

8 Cryptomeria japonica (Japanese Cedar)

9 Malus domestica (Apple skin ethanol extract)

10 Actinostemma lobatum Cucurbitaceae (methanolic extract)

Plant extracts mentioned in above Table 1.2 are few examples among many

others, hundreds of plant extracts have been screened against ACE in past 20-30

years a list of plant extracts along with their activity is given in (Barbosa-Filho et

al., 2006) review article.

Table 1.3: Peptides from different food proteins as ACE inhibitors.

S.No. Source of Peptide Peptide Sequence

1 Corn gluten AY

2 Wheat IY

3 Whey KW

4 Pea, Rice, Soy FW

5 Alpha – casein FY

6 Rice TQVY

Page 37: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

18

7 Lysozyme VAW

8 Sardine GWAP

9 Shrimps DP

10 Egg RVPSL

11 Salmon FNVPLYE

12 Chicken GAXGLXGP

13 Soybean LAIPVNKP

14 Walnut WPERPPQIP

15 Ostrich egg AFKDEDTEEVPFR

16 Squid gelatin GPLGLLGFLGPLSAPGAP

Many other peptides have been extracted and checked for their inhibitory potential

against ACE. IC50 values of above mentioned peptides in Table 1.3 and few others

are given in (Martin and Deussen, 2019) review article.

Page 38: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

19

Table 1.4: Flavonoids and their derivates from natural sources, as active ACE inhibitors.

S.No. Group of Flavonoids

Compound Structure

1 Flavonols Quercetin-3-O-(6’’-galoyl)-galactoside

2 Flavonols Quercetin-3-α-arabinopyranoside

O

OHO

OH

O

O

HO OH

OH

OH

OH

Page 39: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

20

3 Flavonols Quercetin-3-β-glucopyranoside

4 Flavonols Quercetin-3-O-α-(6’’’-p-coumaroylglucosyl-β-1,2-rhamnoside)

Page 40: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

21

5 Flavonols Quercetin glucuronide

6 Flavonols Isorhamnetin-3-β-glucopyranoside

7 Flavonols Kaempferol-3-α-arabinopyranoside

Page 41: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

22

8 Anthocyanins Delphinidin-3-O-sambubioside

O

OO

O

OH

OH

OH

HO

HO

OH

OH

HO

OH

HO

HO

9 Anthocyanins Cyanidin-3-O-sambubioside

10 Flavones Apigenin

Page 42: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

23

11 Flavones Luteolin

12 Chalcones Butein

13 Flavan-3-ols Epicatechin-dimer

Page 43: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

24

A comparative study of many other compounds and plant flavonoids, was

published by (Balasuriya and Rupasinghe, 2011) and (Guerrero et al., 2012). IC50

values of above mentioned compounds in Table 1.4 are also provided in these

articles.

As the search for more potent inhibitors of ACE is continue, the importance of ACE

inhibition is increasing day by day, two of the potent inhibitors which are commonly

used as drug, Captopril and Lisinopril, were used in this study as standard

inhibitors.

1.5 Drug Discovery Process

The process of drug discovery commences by selecting a target macromolecule

which can be an enzyme, cells or cellular organism etc. Activity of the target

molecule should be pathogenic in nature or at least it should be associated with

the disorder or disease. After the selection of a target, next step is to screen

against different classes of drugs or compounds to identify the leads which have

potential activity against the targeted molecules. In the last step ultimately

optimization of that lead discovery as standard drug (de Boer et al., 2007).

Enzymes are undoubtedly one of the most significant class of drug targets. In past

few years, among all the drugs, 28% of them used to target the enzymes, and

among all those small molecules drugs available in marketed, 47% of them are

purposed to inhibit the enzyme’s activity (de Boer et al., 2007). From period 2006-

2011 (6 years) the world drug market received a total of 149 new standard drugs

including 26 biologicals drugs and 123 small molecule drugs, about one-third

(51/149) of these drugs have the purpose of inhibiting the enzyme activity. Among

all those new drugs, If only peptides and small molecules drugs are considered,

then this percentage increases to 41% (51/123). In these new 51 small molecule

drugs, 14 of them were protease enzymes inhibitors (6 for cardiovascular disease,

3 for viral infection and 5 for diabetes), 12 of them were kinase enzymes inhibitors

(10 of them were used for the treatment of cancer), and 11of them inhibit those

enzymes which are involve in ribonucleic acid (RNA) or deoxyribonucleic acid

(DNA) related function or synthesis (8 of them were used for bacterial or viral

infection and 3 of them for the treatment of proliferative syndrome or cancer)

Page 44: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

25

(Silverman and Holladay, 2014). This large proportion of drugs is because of the

important and necessary role of enzymes play pathophysiology and in regulation

of life processes.

Most common reason to use enzymes as a drug target is that many disorders and

diseases are due to the over or under expression and dysfunctional of enzymes;

by inhibition or modulating their activity these diseases can be controlled. Another

motive to concentrate on the enzymes using as drug targets is that, for the

discovery of small molecules as inhibitors is easy as enzymes are relatively easy

targets. The lead inhibitors which are screened against the target may come from

both natural and synthetic sources (Greis et al., 2006; G. Wu, Yuan, and Hodge,

2003).

Fast and robust methods are very crucial step in the process of drug discovery

and development, for the monitoring of enzyme activity and potential of inhibitor.

As the number of new natural and synthetics compounds in increasing at a rapid

rate there for the high-throughput screening (HTS) is very desired to speed up

screening process, for this purpose, new technologies may be used to develop

fast, sensitive and robust method that can broadening scopes of analysis of all

range of compounds and increase the work flow of drug discovery.

In biotechnology, pharmaceutical industries and enzymatic research communities,

chemiluminescent and fluorescent based screening assays are the first choice

since the beginning of these analyses. One reason for this choice is that these

assays are simple and homogenous, and a range of different assays can be

performed by using the same reagents; but the main challenge faced by these

types of methods is to overcome the need of high-speed analyses while reducing

the false negative and false positive readouts which is a common limitation of

these techniques.

Main challenge to fluorescence and chemiluminescence methods is the false

readouts because of the properties of different compound that can increase or

decrease the strength of the signals. Therefore, other similar methods which have

robust readout, high speed, and lower cost by minimizing the reagents which can

further reduce the false results; would be beneficial for the field of enzyme

inhibitors screening. In recent past years the mass spectrometric methods have

Page 45: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

26

continued to advance the speed and sensitivity of assays. Mass spectrometry is

best appropriate for the determination of inhibitory potential and monitoring the

kinetics of enzymatic reactions that results in the difference of mass in product and

substrate. Opportunities to couple advance mass spectrometry techniques with

enzyme assay screening will continue to come to the forefront (Greis et al., 2006;

Kenakin, 2019; Rathore et al., 2008).

1.6 Enzyme Kinetics

1.6.1 Initial Velocity

With the direct quantification of substrate, it is possible to calculate the initiaI

veIocity of the enzymatic reaction which then used for the kinetics study of that

enzyme. In the enzymatic reaction, the concentration of the substrate decreases

rapidly in initial stages, that is when enzyme has high velocity, but after some time

the velocity of the enzyme decreases gradually, as shown in Figure 1.7.

Figure 1.7: Graph for calculation of initial velocity of enzyme reaction.

Page 46: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

27

The initial velocity of the enzymatic reaction can be calculated from the few initial

points of percentage conversion versus time course graph by using the eq. 1.

v� = ∆�∆t (. 1)

Here, vo is the initiaI velocity of enzymatic reaction, ΔC is the difference in

concentration of either the substrate or product at a given time of interval and Δt

is the difference of time for that interval.

1.6.2 Michaelis-Menten Kinetics Vmax and Km

Michaelis-Menten kinetics is used to determine the mode of action of inhibitor on

a particular enzyme, in this kinetics a graph between initial concentration of

substrate [S] and initial velocity (vo) is plotted and inhibition mode is identified by

the different shapes of curve. In this kinetics study Vmax is the maximum initial

velocity of enzymatic reaction which is an indication of enzyme saturation by its

substrate. The Michaelis constant (Km) is that specific concentration of substrate

where enzyme reaction has half of its maximum velocity (Vmax). Km shows the

affinity of a particular substrate to the enzyme. Different types of inhibition have

different values of these kinetic parameter (Vmax and Km), different types of

inhibition are shown in the Figure 1.8 (Balbaa and El Ashry, 2012).

Page 47: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

28

Figure 1.8: Michaelis-Menten kinetics graph and different types of inhibitions.

With comparison to the control reaction, competitive inhibitors do not change the

Vmax but increases the Km., by increasing the concentration of substrate

competitive inhibition can be reduces, because very few active sites will be

available for the inhibitors. On the contrary, the noncompetitive inhibitors decrese

the Vmax but do not change Km value, this mode of inhibition cannot be barred by

increasing the substrate concentration, because both substrate and inhibitor binds

with the different sites of the enzymes. The uncompetitive inhibitions show mixed

types of response because both the Vmax and Km values are changed in this

inhibition (Balbaa and El Ashry, 2012; Silverman and Holladay, 2014). The

inhibition pattern of inhibitors can aIso be interpreted by using the Lineweaver-

Burk & Dixon pIots.

1.6.3 Lineweaver-Burk PIot

Lineweaver-Burk plot is plotted in between the inverse of initial velocity of enzyme

reaction (1/v) and inverse of the concentration of substrate (1/[S]), in the presence

and absence of various fixed concentrations of the inhibitor. For competitive

Page 48: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

29

inhibition the lines of different concentrations of inhibitor [I], intersect on the same

point on y-axis (1/v) which is 1/Vmax, for noncompetitive inhibition lines meet at the

same point of x-axis (1/[S]), which is -1/Km. In uncompetitive or mixed types of

inhibition lines do not intersect and remain parallel to each other with a slope of

slope=Km/Vmax (Palmer and Bonner, 2011).

Figure 1.9: Lineweaver-Burk plots for different types of inhibition.

Page 49: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

30

1.6.4 Dixon Plot

Dixon plots for enzyme kinetic study are plotted in between inverse of the initial

velocity (1/vo) and the concentration of inhibitor [I], while different concentration of

substrate [S] is present. In Dixon pIots, the Iines of different concentrations of

substrate (which have an increasing trend along the horizontal axis) intersect each

other, the point of intersection give the information about kinetics of inhibition, if

the intersection point is between the two axes it is competitive type inhibition, if the

point of intersect is at the x-axis then this type of inhibition is noncompetitive. In

mixed types of inhibition the lines intersect on the y-axis (Palmer and Bonner,

2011).

Figure 1.10: Dixon plots for competitive and noncompetitive inhibition.

Page 50: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

31

Chapter 2: Mass Spectrometry Techniques for

Enzyme Inhibition Analyses

Page 51: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

32

2.1 Introduction to Mass Spectrometry

Mass spectro metry (M S) is a highly precise, reproducible and sensitive technique

which make it ia priority ifor quantitative analyses in comparison to other

techniques. Advances iin mass spectro metry have Ied it to ithe high-throughput

technique of present days as welI as its versatility in hyphenation with other

analytical techniques has made it unique. Mass spectro metry iis used iin several

fields including idrug discovery iand development process, natural product

research, food and environmental sciences, industrial processes, biological

sciences forensics etc.

Mass spectrometry is highly delicate technique, very low amount of sample as low

as in the range of femtomoles (fmol) is required for the analysis, but sample utilized

cannot be recovered as it is destructive in nature. The basic operating principle of

mass spectro metric analysis iis ithe ionization iof ithe sample, transmition of ions into

the mass anaIyzer by appIying high voItage, then separation iof the iions on the

basis of mass- t o- charge ratio (m / z) and finaIIy ithe detection of iions on detector.

Results iof the anaIyses are collated iin the iform of ia spectra, ma ss spectra is a plot

between intensities of different ions on y-axis against their m/z value on x-axis.

Most common information used by the mass spectrum is to find out the mass of

the compounds but rather than that, a lot of information can be extracted form a

mass spectrum. The mass in the spectrum is the sum of monoisotopic masses of

elements not the average masses . This information can be used to determine

the elemental composition of a compounds and thus determine chemical formula.

Fragmentation provides the essential iinformation about ithe structure iof

compounds which ican ibe used for ithe identification of molecules. While the

intensity of peaks can also be used in many different ways, one of the most

common ways is to do the quantification, relative and absolute both, by comparing

with the standard concentrations (Rockwood, Kushnir, and Clarke, 2018).

Generally, ma ss spectro meters have the following components:

1. lon source (EI, CI, DI, FAB, etc.)

2. lon guidance optics (to transfer the ions ito ithe inext istage)

Page 52: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

33

3. Ma ss anaIyzer ( a singIe o r multiple analyzer)

4. Dete ctor (ECD)

Several types iof mass spectro meters are availabIe nowadays iwhich have

icombinations of different iionization isources, iion optics iand mass ianalyzers.

iHowever for general reference ia ischematic diagram iof ian eIectron iionization (EI)

imass spectrometer with magnetic sector anaIyzer is shown in iFigure 2.1.

Figure 2.1: GeneraI schemati c diagram of mass spectro meter (Soderberg, 2019).

2.2 Mass Spectro metry Instrumentation

2.2.1 Ion Sources

The whoIe principle of mass spectro metric anaIysis is stand upo n the ionizatio n of

analyte, since ma ss spectro meters can only detect and analyzed charged particles

(ions). The information obtained from the ma ss spec trum is depends upon ithe itype

of iionization iused. For example, electron ionization (EI) because of its high energy

can produce fragments of molecules even in some cases molecular ion totally

Page 53: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

34

disappeared form the spectra. On the basis of how much fragmentation is

produced in the iionization process, iionization techniques are cIassified as hard

and soft techniq ues. The physicaI iand chemicaI properties iof ithe compound and

the type of information needed, determines the appropriate ionization technique.

Properties of compound which idetermine the ichoice of ionization isource included

moIecuIar weight, voIatility, poIarity and thermaI stabiIity. Ionization methods used

for method developments in this thesis are discussed as follows.

2.2.1.1 Matri x Assis ted Laser Desorption lonization

Mat rix-assis ted Iaser desorption ionization (MALDl) iis relatively similar to the fast-

atomic bombardment (F A B) in terms that it uses a matrix to assists in the ionization

process, but in FAB, ionization take place by the high energy beam of atoms; while

in MALDI the ionization process uses a high energy laser pulse. Common matrices

of MALDI included 2 , 5-dihydr oxy benzoic acid (D HB), sinapic acid, α -cyano-4-

hydro xycinnami c acid (α-CHCA) and feruIic acid. Figure 2.2 is the schematic

representation iof iionization process takes pIace in MALDl ma ss spectrometer.

Figure 2.2: Sche matic diagram of ionization source of MALDl mas s spectrometer (Szot and Croxatto, 2019).

GeneraI method ifor ithe ipreparation iof sample for MADLI anaIysis is to mixed the

analyte with a solution of matrix and spot samples on a stainless-steel plate

specialized made for sample spotting called as MALDI target plate (MTP) then

spots aIIowed to dry iover ithe isurface. A laser pulse of high energy, generally from

ia initrogen (N2) Iaser (3 3 7 nm) o r frequency tripIed and quadrupIed Nd : Y AG Iaser

Page 54: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

35

(3 5 5 and 2 6 6 n m), irradiate the matrix surface and create localized heating which

resuIts in the abIation and desorption iof ithe imatrix along with anaIyte particIes into

the vacuum. The ihot gaseous pIume produces from the abIation process icontains

deprotonated iand protonated anaIyte moIecules which are then guided, through a

potentiaI difference, to the anaIyzer of mass spectrometer where these iions are

separated ion the basis of itheir mass-to-charge (m / z) vaIues then afterwards

detected on the detector.

FAB can be used for the analysis of those analytes which have a mass up to 10

kDa, while MALDl has ian increased range of mass ithat can be extends iup to 500

kDa. Therefore, this technique is very useful for the anaIysis of Iarge biomoIecules

such ias DNA, RNA, prot eins, synthetic polymers an d enzymes. Laser in th e

MALDI pro duces the iions iin puIsed mode therefore, it is usuaIIy equipped with

time-of-fIight (T O F) anaIyzer. This iis ibecause ia TOF anaIyzer ican iseparate

bundIes of iions ithat iare produced at a time ithrough puIsed operation iof ithe Iaser.

In ionization process of MALDI usually a transfer of protons take place between

the molecules of analyte and matrix. The selection of the matrix iis typically depend

upon the type of analyte is being analyzed, for those anaIytes which are acidic in

nature, slightly basic matrix is best choice; while matrices of acidic nature, have

their best use in general applications. Ionization process of MALDI usuaIIy

produces singIy charged ions which are most commonIy protonated i(H+) adducts

ibut isometimes sodium (Na+) iand ipotassium (K+) adducts ican ialso ibe formed (Lee

et al., 2016).

2.2.1.2 Smartbeam Technology

This work iwas iperformed ion the Bruker UItrafIex-IlI, MALDl-T O F/ T O F mass

ispectrοmeter which is iequipped iwith smartbeam Iaser itechnology. Smartbeam

Iaser technοlogy provides significant enhancement in performance of MALDI-MS.

iSmartbeam icοmbines ithe ispeed iof a sοIid-state-Iaser with ithe iwide irange iοf

appIicatiοns iassοciated iwith nitrοgen Iasers. iThe use iof cοnventionaI iNd:YAG

(neodymium-doped i yttrium i aluminum i garnet; i Nd:Y3AI5O12) Iasers is irestricted

itο few sampIe types but ismartbeam iprοvides exceIIent iresults frοm aII imatrices

Page 55: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

36

iand aII sample ipreparation imethοds. A comparative spectra of conventional and

smartbeam source is given below in Figure 2.3 (Holle and Ketterlinus, 2008).

Figure 2.3: Comparasion of Nd:YAG laser and smartbeam spectra (Holle and Ketterlinus, 2008).

Smartbeam use the ifrequency-tripIed iNd:YAG Iaser, iat i3 5 5 n m and a istructured

ifοcus prοfiIe, i‘Nd:YAG i(structured iA) and iNd:YAG i(structured iB), it allows

iswitching iamοng iN2, iNd:YAG i(Gaussian) & iNd:YAG i(structured iA ior iB) ilasers

iwithin isecοnds, schematics of Smartbeam setup are shown below in Figure 2.4

(Holle et al., 2006).

Figure 2.4: Smartbeam setup (Holle et al., 2006).

Page 56: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

37

Smartbeam used in the instrument had following characteristics: Po ≤ 30 mW, Pp

≤ 30 kW, t = 5 ns -20 ns, F = 200 Hz (single impulse), λ = 990-1080,808,495-

540,330-360 nm

2.2.1.3 EIectrospray lonization

EIectrospray ionization (ESl) is an atmospheric ipressure iionization (A PI)

techniqu e, it is used for the wide range of analyses and various types of analytes.

It was 1st deveIoped by Masamichi Yamashita and John Fenn in 1984 (Yamashita

and Fenn, 1984). ESl comprised of process, in which anaIyte particIes are

transferred from soIution phase to th e gas phas e through soIvent or i on

evaporation (Somogyi, 2008). ESI is a soft ionization technique it usualIy produces

very Iittle or no fragmentation and it can be very easily hyphenated with Iiquid

chromatographic techniques. Among other advantages a major benefit of ESl is,

the generation of ions with multiple charge, which decrease the m/z of ions

therefore, increases the mass range of the anaIyzer and makes it suitable for the

anaIysis of molecules with very high moIecuIar weight.

Process of ionization needs dissolution of analyte in poIar solvent, then soIution is

nebuIized through spray from a capiIIary that is kept at a very high potentiaI with

a capilIary voItages of 3 to 6 kV. The dropIets of Iiquid attain charges from the high

voltage, when desolvation process take place in the droplets, solvent is removed

from the droplets by the action of ihot icurrent iof drying gas. Because iof this drying

iprocess droplets starts to ishrink which increase the columbic repulsion between

ions. Droplets shrunk up to some extent after that when surface itension iof ithe

soIvent ican no Ionger abIe to hoId the couIombic repuIsions and iits reached at its

Iimit which is called as Rayleigh limit (Rayleigh, 1882). After that, shrunk dropIets

burst and reIease the ions in the iionization ichamber which are then moved to the

anaIyzer region by applying a potential difference. iFigure 2.5 demonstrate the

process of desolvation and ionization in ESl.

Page 57: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

38

Figure 2.5: Process of desoIvation and ionization in ESl-MS (IPFS).

In hyphenation of liquid chromatography (LC) with mass spectrometry, ESI-MS is

given the priority, and this hyph enation make s it a hi gh-throughpu t te chnique.

Ma ss spectro meters utiIizing the ESl source are generally equipped with a

combination of mass anaIyzers; the most common combinations are quadrupoIe

time-of-fIight (Q qT OF) and triple quadrupole (QqQ). These mass anaIyzers

enabIe sensitive quantitat ion of differen t anaIytes in compIex sampIes (Cífková et

al., 2015; Mirzaian et al., 2017), iidentification of inatural products from pIant

extracts (Abu-Reidah et al., 2015; Allen, Greiner, and Wishart, 2015) and

screening of enzymatic assay against drug libraries by using absolute

quantification of substrate and peptides (Cui, Nithipatikom, and Campbell, 2007).

Formation of multipIy charged iions in ESl make iit possible ito analyze the large

molecules like nucleic acids and proteins on quadrupoIes whose imass range iis iup

to 2 0 0 0 D a (Percy et al., 2016; Ruhaak, 2017).

2.2.1.4 Agilent Jet Stream Ion Source

Jet Stream iοn sοurce with thermaI igradient ifοcusing technοIogy iby AgiIent,

οptimizes iESI iconditions to iproduce significant igains in isensitivity, idecreasing

sampIe isize, iincreasing sampIe ithrοughput, iimprοve irοbustness iand ireduce ithe

LOD i& iLOQ of iscreening and quantitatiοn iappIicatiοns iby imprοving ithe

Page 58: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

39

desοIvation and ispatiaI fοcusing of iiοns Figure 2.6. Superheated initrοgen isheath

igas icοnfines ithe nebuIizer ispray itο imοre effectiveIy idry iiοns iand cοncentrate ithem

in ia thermaI icοnfinement zοne. idesοIvation reduces inοise in ispectra Figure 2.7

(Mordehai and Fjeldsted, 2009).

Figure 2.6: Jet stream ionization source working principle (Mordehai and Fjeldsted, 2009).

Figure 2.7: Thermal profile of jet stream source showing confinement of zoning by super-heated nitrogen (Mordehai and Fjeldsted, 2009).

Page 59: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

40

FuII cοnfinement οf the ispray iby ithe isuperheated nitrοgen igas eIiminates the

sampIe recircuIatiοn and ireduces ithe ipeak taiIing. It iimproves the iprοductiοn iοf

iοns which resuIts iin ihigher intensity οf peaks in spectra and improved signal-to-

noise ratios. Jet stream source can improve the MS and MS/MS spectra intensity

5 to 10-fold, Figure 2.8 (Mordehai and Fjeldsted, 2009).

Figure 2.8: Comparison of spectra using conventional ESI and jet stream technology (Mordehai and Fjeldsted, 2009).

2.2.2 Mass Analyzers

The mass analyzer is the main component of any mass spectrometer that perform

the function of separation of ions, on the basis of their mass-to-charge (m/z) ratio.

Different kinds of mass analyzers have different operating principles, as an

example, the magnetic sector, oldest mass analyzer, works upon the principle of

magnetic deflection, the ions deflects as they travel through it and detect on the

detector. Separation is based on how much an ion is deflected under the applied

magnetic field. There are six types of mass analyzers as listed below.

1. Electrostatic sector (E)

2. Magnetic sector (B)

Page 60: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

41

3. Quadrupole (Q)

4. Time-of-flight (TOF)

5. Ion traps including the orbitrap

6. Ion cyclotron resonance (ICR)

However, only those analyzers are discussed here which are used in this study.

2.2.2.1 Quadruple Mass Analyzer

A quadrupole (q) mass analyzer is comprised of four metallic rods, aligned in a

parallel manner. The two of opposite facing rods are connected to a direct current

(DC) source and other two are connected to the radiofrequency (RF) which is at

an offset to the DC voltage. The quadrupole works as an tunnel for ion-

transmission where, at specific DC and RF voltage, only selected ions with certain

mass-to-ratio (m/z) can pass by forming a stable trajectory, while other ions which

have unstable trajectories, cannot pass through and are destroyed by colliding

with the quadrupole rods. The working principle of a quadrupole mass analyzer is

illustrated in the Figure 2.9.

Figure 2.9: Transmission of different ions through quadrupole mass analyser (Khan, 2019).

Page 61: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

42

The quadrupole mass analyzer is the most typical type of analyzer available in

low-cost mass spectrometers because if its robustness, ease of manufacturing,

and fast speed of scanning. The speed of quadrupole mass analyzer is compatible

with fast liquid chromatography separations which can produce peak narrower

than 5s. The quadruple mass analyser has low-resolution unlike the magnetic

sector and time-of-flight (TOF) analyser which have high resolution capacity. It is

therefore impractical to deduce the exact molecular weight and formulas by using

the data generated from a quadrupole instrument, instead, quadruples have

proved to be best in quantification studies.

2.2.2.2 Time-of-Flight Mass Analyzer

Time-of-flight (TOF) mass analyzer operates on the principle that, ions with

different molecular weight but same values of kinetic energy will have different

velocities in a field free region. In TOF, packet of ions produced in the ionization

chamber is accelerated to a specific kinetic energy and moved to the TOF analyzer

where heavier ions have slower speeds as compared to the lighter ions. Those

ions which have samller m/z values will reach the detector first followed by the

ions which have higher m/z values. Figure 2.10 shows the separation of ions

based on their difference in velocities in a TOF mass analyzer.

Figure 2.10: Ions separation as a function of time in a TOF analyzer.

Page 62: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

43

TOF has a high resolution; newer TOF analyzers can reach up to the resolution of

80,000. Different makers use different variations in TOF analyzers; one of the

advantageous variations is using of reflectrons to increase the travel distance of

ions two or more times, the longer the ions travel, the greater will be the resolution.

Some manufacturers use longer path TOF tubes to avoid the use of reflectrons

which may reduces the cost of manufacturing.

One limition of the TOF analyzer is that, it cannot work in a mode where ions are

produces continuously, it can only be used with the source which produce the ions

in packets. it will become very difficult to monitor the time of flight of different ions

if ions are continuously injected into the analyzer. Therefore, TOF is most

compatible with MALDI instrument where the ionization source produces ions in a

discontinuous manner.

Unlike the quadrupole mass analyzer, a TOF analyzer is unable to select a specific

ion of certain m/z value, all ions will pass and rech the detector or next phase

ultimately. Therefore, TOF instruments are unable to work under some specific

modes such as the multiple reaction monitoring (MRM), but QTOF and QqTOF

are the tandem techniques which are generally used. TOF are not well suited for

quantitation purposes but can give high accurate mass which can be useful for

determination of composition and structure of an analyte.

2.2.3 Detectors

Detector is the final part of a mass instrument, ions produced in the ion source

then separated by the mass analyzer finally reached at the detector and generate

the signal which then converted into the data. The purpose of the detector is to

produce a signal in the form of electric current which corresponds to the

abundance of ions hitting the detector. This information later converted into useful

data in the form of a mass spectra. If the ion intensity of an analyte is very low,

then amplification can be used on detectors.

Choice of detector is dependent upon the intended application and design of the

instrument. The detection of ions is based on mass, charge and velocity of ions.

Different detectors detect ions in different manner; for example, Faraday cups

Page 63: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

44

detectors record the current generated directly when an ion hits the detector

surface, while electron multipliers detector record an amplified current that is

produced due to secondary electrons generated in the amplification process.

Detection principle in Fourier-transform ion cyclotron resonance (FTICR) mass

spectrometers is entirely different from all other types, because in FTICR there is

no need of a separate detector, but the detection takes place in form of induced

current when ions within the mass analyzer pass close to the electrodes.

2.3 Tandem Mass Spectrometry

Tandem mass spectrometry is the combination of at least two stages of mass

analysis by the combination of two or more mass analyzers. Such types of

instrument are known as hybrid instruments and quadrupoIe time-of-fIight

(Q q T OF) and tripIe quadrupoIes (Q qQ) are examples of it. Trapping instruments

works in different way, they only have one analyzer, yet ithey ican pe rform tan dem

ma ss spectro metry e xperiments. Based on how mass anaIysis take pIace in,

tandem mass spectrometry is classified into two types; tandem- in -space and

tandem- in -time.

2.3.1 Tandem-in-space Mass Spectrometry

During tandem-in-space, ions traveI some distance in phases of selectin,

fragmentation and detection, it is usually observed in the sect or, quadrupIe and

T O F mass analyzers. TripIe quadrupoIes is the most com mon combination of

mass anaIyzer in hybrid instruments and it provides the most variety of scan types.

2.3.2 MS/MS by TripIe QuadrupoIes

Triple quadrupole is comprised of three quadrupoIes, in which first and last

quadrupoIes are used for selection or scanning on ions while the middle

quadrupole serves as the colIision ceIl where ion fragmentation takes place. A

schematic diagram of a tripIe quadrupoIe is shown below in Figure 2.11. Collision

energy required for the fragmentation of ions is determined by the kinetic energy

of ions arriving from the first quadrupoIe, if there is no requirement of

Page 64: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

45

fragmentation it ican ialso iwork ias ia transmission device. Diffe rent working modes

of tripIe quadrupoIes are summarized as follows:

1. Produc t-io n sc an

2. Precurso r-i on s can

3. SeIected i on mo nitoring (SIM)

4. SingIe and muItiple reaction monitoring (SRM / MRM)

Figure 2.11: Sch ematic of a tripIe quadrupoIe ma ss spectro meter (Jackson, 2019).

2.3.2.1 Product -lon Scan

In product-ion-scanning, the Q1 is set at specific m/z value to allow only certain

ions (within the toIerance window) can pass through, which are th en fragment ed

in the reaction ceIl q2 and finally the product io ns formed in the resuIt of

fragmentation, are anaIyzed in the Q3, which is set ito iscan a ispecific irange of m/z

values. This type of scan iis used to idetermine which ifragments resuIt from ia

cert ain prec ursor iion and iis used for dete rmination iof fragm entation ipatterns of

icompounds.

2.3.2.2 Prec ursor-lon Scan

Precursor-ion scan is reverse of product-ion-scan, in this type of scan, Q 1 is set

to scan a specific range of m/z values, q 2 is set to make f fragments and Q 3 is set

on a specific m/z value. A signaI wilI generate only when a precursor ion from Q 1

Page 65: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

46

produces a frag ment at which Q 3 is par ked. It can also ibe iused ifor the istudy of

ifragmentation pattern.

2.3.2.3 SeIected-lon Monitoring

In seIected-ion monitoring (SlM)., both Q 1 and q 2 are turned off and onIy serve to

transmit the ions to Q 3, while Q 3 scans over the set range of m / z vaIues. This type

of scan is generaIly used for quanti tative anaIysis in a simpIe mixture, where it can

give good sens itivity. In the case of Iarge and compIex mix tures, there is a strong

possibiIity to observe iso baric ions that eIute at the same reten tion times.

2.3.2.4 SingIe and MuItiple Reaction Monitoring

Single or selected reaction monitoring (SRM) is a neutral loss scan. In this

scanning both Q1 and Q3 are set on a specific m/z values and q2 is served as

reaction cell for fragmentation. Ions which passes through the Q1 are precursor

ions and in Q3 these are fragment ions. Before setting the values in Q1 and Q3, a

SIM for high intensity of precursor ion and product ion scan for fragments can be

done to select the ions which have highest intensity to increase the sensitivity of

analysis.

MuItiple reaction monitoring (MRM) is just two or more SRMs taking pIace

simuItaneously. MRM can anaIyze two or more compounds in the same

experiment as welI as in M R M two or more product i ons from a singIe precurs or

ion can also be anaIysed. In M R M experiment, Q 1 and Q 3 do not scan the whoIe

range of m / z; instead they jump from one m / z vaIue to another. Jumping instead

of scann ing enhances the duty cycle of the instrument a nd increas es t he

se nsitivity.

2.3.3 MS / MS by Qq TOF Sy stems

QuadrupoIe-time-of-fIight hybrid syste ms are different from the tripIe quadrupoIe

systems iin the way ithat, Q 3 iis repIaced by a T OF mass anaIyzer. T OF has very

high resoIution scanning capabiIities, a Qq T OF system ca n record high resoIution

mass spectra of precursor iions when no fragmentation iis performed and iof product

ions iwhen fragmentation iis performed.

Page 66: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

47

These iinstruments are of great vaIue when structuraI iinformation iis required

through fragmentation istudies. UsuaIIy, a TO F scan is performed first iwhere Q 1

and q 2 iare not scanning and onIy act as itransmission devices and ithe T OF

anaIyzer gives ithe exact masses of aII produced iions. In the MS/MS scan the

quadrupoIe iis used to seIectiveIy fiIter iions of iinterest which are then ifragmented

inside q 2 and the product ions iare transmitted to the TOF region where they are

separated. QqT OF systems can record mass spectra at significantIy higher

resoIution than the Q qQ systems but are unabIe to perform M RM scans as the

T OF anaIyzer is fieId-free and cannot perform ion seIection.

2.3.4 Tandem-in-time Mass Spectrometry

ln tandem-in-time mass spectrometry, iions do not traveI through different mass

anaIyzers, instead, they remain iconfined iin a singIe mass anaIyzer during aII the

stages iof mass spectrometry. lt is usuaIIy performed iin trapping iinstruments and

does not require a hybrid iinstrument; as a singIe ion itrap can store, fragment iand

separate iions. Different types of mass anaIyzers used iin tandem-in-time mass

spectrometry are:

1. QuadrupoIe lon Trap (PauI Tr ap)

2. Orbi trap

3. Penn ing Trap (us ed in FTlCR-M S)

lnstruments equipped with ion traps are not Iimited to M S 2 or M S 3 as the Qq Q and

Qq TOF systems but are abIe to perform muItipIe stages of ion s eparation and

fragmentation. Modern instru ments can go as high as MS 1 1. Such instruments can

be used to extract vaIuabIe structuraI infor mation.

2.3.5 Fragmentation and Ion Activation

In tandem mass spectrometry, fragmentation is an important step. Hybrid

instrum ents have coIIision / reaction ceIIs for i on fragment ation whiIe an iion trap

does inot require a iseparate reaction ceIl iand can perform ifragmentation iitself.

Different methods iof fragmentation are iused incIuding iln-source iFragmentation,

CoIlisionaI-induced iDissociation (Sleno and Volmer, 2004), which can be iLow

energy ClD and High Energy ClD (Sleno and Volmer, 2004).

Page 67: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

48

There are other dissociation techniques like; EIectron Cap ture Disso ciation (E C D),

EIectron T ransfer Disso ciation (E T D), Photodissociation (PD) (Mistarz et al.,

2018), Surface Induced Dissociation (SID) (Yan et al., 2017), lnfrared MuItiphoton

Disso ciation (IR M PD) (Brodbelt and Wilson, 2009) and BIackbody lnfrared

Radiativ e Disso ciation (BI RD) (Dunbar, 2004; Sleno and Volmer, 2004).

2.4 Mass Spectrometry for Enzyme Assay and Inhibitors

Screening

Applications of mass spectrometry have been developing and expanding since

last two decades due to the advancement and development of instrumentation,

mass spectrometry have many advantages for the fieId of inhibitor screening. The

main reason iis the abiIity to perform iscreening assays in a IabeI-free mode. iIn

contrast to uItra vioIet, fIuorescence and iradioactivity assays, detection iof

enzyme-inhibitor iinteractions does not irequire IabeIing but reIies entireIy on ithe

mass-to-charge ratio iof reporter moIecuIes. This simpIifies adjusting the iassays,

which are no Ionger restrained by the ineed to design and synthesize IabeIed

compounds. LabeI-free assays aIIow Iaborious, cumbersome iand costIy

derivatization ito ibe ibypassed (de Boer et al., 2007).

In mass spectrometry it is possible all types of compounds compared to only one

if label-based detection like fIuorescence is used. It means that, by applying mass

spectrometric method, all components of an enzyme assay (e .g., subs trate,

products, cofa ctors, coenzymes, and internaI standar ds) can be us ed t o monitor

the reIative enzyme activity or inhibition (Liesener et al., 2005). Furthermore, the

identification and structural determination can also be done at the same time by

using the accurate molecular mass of the inhibitor by T OF-MS or FT-lCR-MS) in

combination with fragm entation (M S / M S) experiments. Finally, the main

disadvantages associated with the fIuorescence based assays is the false

enhancement or decrease of results by means of auto fIuorescence or light

scattering o f the compound s or matrix, and quench ing; which do not exist for MS.

This may reduce the faIse positives and faIse negatives resuIts (de Boer et al.,

2007).

Page 68: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

49

MS readout aIso offers a significant advantage. For exampIe, the reagent cost for

a MAIDI-Qq Q readout is onIy ~$ 0.0 3 per weII for the buffer and the unIabeIed

peptide substrate compared with $ 0. 60 per weII for A D P accumuIation assay.

AIthough this may not seem aII that significant when measuring onIy a few test

vaIidation sampIes, when scaIing up to evaIuate a compound repository of

10 0,000 compounds, the reagent cost difference becomes $ 3000 for the MS

versus $ 6 0,0 00 for the fIuorescent readout. At this rate of saving in reagent costs

aIone, the investment cost for the mass spectrometer ($ 3 50,000 to $ 45 0,000)

couId be recovered after onIy a short time by a productive screening group. Thus,

with comparabIe readout speeds, better signaI-to-background ratios, a significant

reagent cost advantage, and the IikeIihood of fewer faIse-positive or faIse-

negative resuIts, the MALDl-Qq Q readout represents a major step forward for the

H TS readout of isoIated enzyme reaction (Rathore et al., 2008).

2.5 Methods of Mass Spectrometry for Enzyme Assay

Screening

The Mass Spectro metry based screening methods are not only Iimited to the

analysis of enzyme-inhibitor reactions, but they can be used for receptor-ligand

too. Receptors are those proteins which are present on celIs or within the

cytopIasm or ceIl nucIeus that bind to receptors (specific compounds). Enzymes

are the analogues of receptors and inhibitors are analogues of ligand. In this thesis

research is being done by using enzyme and inhibitors. Since there is a strong

overIap in the methodologies used for both enzymes and receptors screening;

methodologies which are discussed further can be used for both types of target.

The mass spectrometric methods used for the screening of enzyme can be

cIassified according to differ ent criteria, such as the drug target, type of assay,

physicaI use of drug targets, the type of sampIe/inhibitors screened, or the type of

mass spectrometric techniques. ln further discussion, described methodoIogies

are categorized on the basis of detection principIe i.e. which component of the

enzyme reaction is to be detected. Three categories of mass spectrometry based

screening are also shown in Figure 2.12.

Page 69: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

50

Figure 2.12: Different types of mass spectrometry based screening assay (de Boer et al., 2007).

Based on which component is detected in enzyme assay; screening is categorized

into three methods i.e. detection of enzyme inhibitor complex, detection of inhibitor

and detection of reporter molecule, substrate or product, among them first and

second are the direct approach to measure the enzyme reaction while third one is

indirect way.

Direct screening methods detect ithe active icompound(s) either as iintact enzyme -

inhibitor compIex or iafter dissociation iof the enzyme - inhibitor compIex. In

detection of enzyme-inhibitor complex, mass spectrometer detects the cumulative

mass of enzyme with inhibitor and detect the inhibitor based on mass difference.

In detection of inhibitor it only determines the mass of inhibitor after the

dis sociation of enzyme - inhibitor compIex, in this scre ening method separation of

bounded compounds from the unbounded is necessary to differentiate the active

compounds from the inactive. For indirect screening methods, substrate or product

of enzymes are used as the reporter moIecuIes to measure the enzymatic activity.

Usually in indirect screening methods it is not needed to separate the inhibitor or

reporter molecule from the assay soIution after incubation, they can directly

analyze by choosing appropriate reporter molecules, but a separation step can

Page 70: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

51

enhance the sensitivity. In most cases, the concentration of the reporter moIecuIe

is directIy reIated to the activity of enzyme. In most of the enzyme assays, it is

easy to detect the products formed in the enzyme reaction; in those case, a

decrease in the concentration of product or increase in the substrate at a given

time can give the signal of inhibition of enzyme (de Boer et al., 2007).

Various mass spectrometric techniques used for above describes methods of

detection are used, which are describes in the Figure 2.13. each technique has

its pros and cons, advantages over the other. Over the time as the instrumentation

is improved in speed and sensitivity more reliable and accurate methods are

developed for the enzyme assay screening.

Figure 2.13: Different techniques of mass spectrometry used for enzyme assay screening.

2.5.1 Direct Infusion-Mass Spectrometry (DI-MS)

This technique of enzyme inhibitor complex is very simple, it was made easy by

the using of electrospray ionization (ESI). Analysis of high molecular weight

compounds in range of KDa like enzyme is relatively easy on ESI due to

Page 71: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

52

accommodation of multiple charges on the substance. In this technique the

inhibitor or mixture of inhibitors incubate with the enzyme and after that it subjected

to the MS analysis, the mass spectrum showed the multiple charged enzyme -

inhibitor compIexes, which demons trate the bind ing betwe en inhibitor a nd

enzyme, an ESI spectra of enzyme inhibitor complexes is shown in Figure 2.14

(Baca and Kent, 1992; Stevenson, Feng, and Storer, 1990).

Figure 2.14: Direct Ionization mass spectrometry spectra of enzyme inhibitor complexes (Stokvis et al., 2000).

The direct detection method can also be used to determine the relative binding

affinities of inhibitors by using ESI-MS. Stokvis et al. demonstrate the reliability of

MS detection for enzyme-inhibitor complexes and determination of binding

affinities of inhibitors against carbonic anhydrase. In that methods, screening was

much faster and required very less reagents than spectrophotometric analyses.

Moreover, this MS based method could be used when concentration of inhibitor is

high, which is unfavorable for spectrophotometric analyses (Stokvis et al., 2000).

Direct screening method of enzyme-inhibitor comples, is not only limited for the

analysis of individual compounds but it can also be used for complex samples

(Sannes-Lowery et al., 2004) or compound libraries. In an example demonstrated

by Gao et al. that, a library of 28 9 pep tides was incub ated with carb onic an hydrase

II enzyme and then, that mix ture was anaIyzed by eIectro spray ioniza tion fo urier

trans form i on cycIotron reso nance ma ss spectro metry (ESl-F T-lCR-M S). Because

of high resolution, F T-lCR- M S can isoIate and accumuIate enzyme-inhibitor

compIexes. This method can also used for the both bound and unbound analysis,

Page 72: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

53

after dissociation of enzyme-inhibitor complexe unbound compounds can be

detected and by comparing the relative abundances of the detected inhibitors with

the abundances of the compIexes, binding constants of the inhibitors can be

calculated Figure 2.15. The correIation between abundance and binding

constants can be made onIy if the ionization efficiencies of aIl compIexes are

simiIar (J. Gao et al., 1996).

Figure 2.15: ESl-FT-lCR-MS screening o f library of peptides (J. Gao et al., 1996).

Page 73: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

54

This technique is advantageous that it can be used to detect all the components

of an enzyme reaction, the ratio of dissociate to undissociated can be used to find

the binding efficiency of inhibitors. High resolution of FTICR make it possible to

detect closely mass peptides and complexes. While on the other hand this

technique has low tolerance for the high concentration of buffers, salts,

surfactants, co-factors, etc., in this case sensitivity may be increase by using some

pretreatment methods. The detection of inhibitors has advantage over the

detection of enzyme-inhibitor complex, beca use in enzyme-inhibitor complex due

to very high mass and multiple charges the difference in the mass will be very

small.

2.5.2 Ultrafiltration-Ma ss Spectro metry (UF-M S)

Enzymes and inhibitors have huge differencea in their size, ultrafiltration-mass

spectrometry (UF-MS) use this difference in the size to perform the analysis. UF-

MS utilize a fIow- through vessel for perfoming enzyme assay, that vesseI contain

an uItrafiItration mem brane with a defined molecuIar-w eight cut- off. The

membrane traps the enzyme moIecules or enzyme inhibitor compIex but aIIows

the Iow moIecular weight compounds to pass through, as shown in Figure 2.16.

During enzyme assay screening, the target enzyme is incubated with the different

inhibitors in the mixing chamber, after incubation its components are separated by

the UF membrane. The enzyme-inhibitor complexes or free enzymes are retained

of the UF, while the unbound compounds are removed by washing the retentate.

After the washing step, the complexes are dissociated by using an organic modifier

or by changing the pH of solution, or by using a combination of both. Released

inhibitors are then transported through the membrane and analyzed by mass

spectrometer, by comparing the spectra of all inhibitors before and after the

incubation, inhibitors which bound with the enzymes can easily be identified

(Johnson, Nikolic, and van Breemen, 2002; van Breemen et al., 1997; Youngquist

et al., 1995; Zhao et al., 1997).

Page 74: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

55

Figure 2.16: Ultrafiltration process in enzymatic assays (de Boer et al., 2007).

UF-MS techniq ues ca n b e us ed for the compIex mix tures like extracts of plants or

microorganism (Zhang et al., 2004), as shown by the Wang et al. in Figure 2.17,

that, ultrasonic extract of herb Scutellaria baicalensis was incubated with α-

Glucosidase enzyme which is a contributing factor to diabetic mellitus type-II, then

unbound compounds were separated by the mixture in a cartridge filter contacting

UF membrane by using the centrifugation, after the washing, compounds were

released by using a organic solvent and released compounds were analyzed on

ultrafiltration liquid chromatography with photodiode array detection coupled to

electrospray ionization tandem mass spectrometry (UF-LC-PDA-ESI/MS) (J.

Wang et al., 2014).

Figure 2.17: α-Glucosidase inhibitory assay with S. baicalensis extract using UF-LC-PDA-ESI/MS (J. Wang et al., 2014).

Page 75: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

56

By using the HPLC prior to MS can make a screening assay more sensitive,

selective and efficient as well ionization suppression in the MS can also reduce by

HPLC. Dissociated inhibitors cane be separated before injected into the MS. In

addition, the HPLC.

In another example Nikolic et al. demonstrate the potential of UF-MS for screening

of natural products. The fermentation-broth extracts and plant extracts along with

standard inhibitors were screened against COX-2 enzyme. The incubation mixture

was injected into the UF chamber with a 30,000-MW cut-off membrane and

washed with water. After that, bonded inhibitors were released by lowering the pH,

then released inhibitors were separated by RP-HPLC and analyzed by MS.

Results showed that three known COX-2 standard inhibitors (ibuprofen, dicIofenac

and fIurbiprofen), labeled 1–3 in Figure 2.18, were present in the spectra along

with peaks of other inhibitors from the plant and broth extracts. Presence of

standard inhibitors in results validated the method and proved the efficiency of UF-

MS even in complex matrices. Besides, the authors showed that extensive

washing wiIl resuIt, as expected, in the Ioss of compounds with weaker affinity

(Nikolic et al., 2000).

Figure 2.18: UF-MS analysis of COX-II inhibitory assay (Nikolic et al., 2000).

Ultrafiltration can also be used with MALDI-TOF-MS. UF is advantageous over the

direct infusion techniques because it uses washing step which can remove the

salts and buffer from the mixture. In UF technique enzymes can be reuse for

multiple cycle, however, if extensive washing is used, may degrade the enzyme

activity. UF does not required the immobilization of enzyme, but if enzyme got

stuck inside the UF membrane then it can be problematic.

Page 76: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

57

2.5.3 Size-Exclusion Chromatography-Mass Spectrometry (SEC-

MS)

Size-excIusion chromatograph (S EC) aIso use the advantage of difference in the

size of enzyme and its inhibitors, in SCE enzyme-inhibitor compIex are separated

from the unbound compounds by using a coIumn. In S EC-M S screening is

performed by incubating the enzyme with inhibitors and injecting this mixture into

a size-excIusion coIumn to separate the enzyme-inhibitor compIexes from the

unbound compounds i.e. separation of high moIecuIar weight components from

Iow moIecuIar weight on the basis of different retention voIumes. SEC work on the

principIe that, moIecuIes Iarger than the pore sizes of the coIumn materiaI cannot

enter the pores therefore they eIute together in first, whiIe the smaIIer compounds

which can penetrate into the pores wiII eIute Iater. The earIy eIuting enzyme-

inhibitor compIexes are isoIated, dissociated by using change in pH or organic

modifier, or temperature change, or a combination of these parameters and eIuent

is finaIIy anaIyzed by the mass spectrometry (Mathur et al., 2005).

In an example, Annis et al. used online hyphenated SEC-MS, in which enzyme-

inhibitor complex after passing through SEC were trapped on HPLC column, and

after washing and dissociation, analyzed by HPLC-MS. With this method, a library

of 2500 compounds were screened against the enzyme Esche richia co Ii

dihydro foIate reduct ase, within 1 0 minutes. Rapid separation of SEC make it

possibIe to anaIyzed the even weakIy -bounded inhibitors with moderat e

dissociation rates and to identified them as possibIe Iead structures for inhibition

of target enzyme (Annis et al., 2004).

Page 77: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

58

Figure 2.19: Fast SEC-MS of a library of compounds and identification on MS/MS basis (Annis et al., 2004).

In the above Figure 2.19 accurate masses and MS/MS information is provided,

which resulted in the identification of a various active compound. Spectra A shows

the EIC of an inhibitor with m/z 515.24. Spectra B is the EIC at m/z 515.24 in a

blank experiment, where no inhibitor or library compounds was used, therefore,

contained no peaks at specified m/z. When enzymatic assay was performed by

SEC-MS/MS, enzyme was incubated with the library, complex was separated,

unbounded and analyzed in MS/MS. Chromatogram at C showed the presence of

compounds at m/z 515.24 and when subjected to MS/MS it results in mass spectra

showed at D. To identify and verify the structure of the compound with m/z 515.24,

different position isomers were prepared independently, and their MS/MS

experiments were carried out, spectra of two of the isomers are shown in the E &

F. By comparing the MS/MS spectra of compound at D with the isomers it was

confirmed that this compound is similar to sub-library.

Many researchers have shown the potential of SEC-MS by adding different

modifications and techniques (Davis, Anderegg, and Blanchard, 1999;

Dunayevskiy et al., 1997; Moy et al., 2001; Siegel et al., 1998; Wabnitz and Loo,

2002) like, use of spin coIumns, which uses the centrifuge to perform the

Page 78: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

59

separation step. The use of spin column in 96-well plate format by

(Muckenschnabel et al., 2004) increase the assay speed for screening up to

85,000 compounds per day. SEC-MS is advantageous in a way that all the

components of enzymatic reaction can be analyzed by this technique as well as

the comparative analysis is also possible by comparing the free ligands with the

initial spectra of library. In SEC equilibrium dissociation may cause loss of some

ligands in column.

2.5.4 Immobilized Enzyme-Mass Spectrometry (IE-MS)

Enzyme immobilization means that the enzyme molecule is attached to a solid

surface without losing their functionality and attachment should be tight enough to

prevent dissociation of the enzymes from the surface (Sirisha, Jain, and Jain,

2016). Enzyme immobilization can be defined as, restricting the enzyme

molecules to a solid matrix/support different from the one in which substrate or the

products are present. It is achieved by attaching the enzymes to or within some

suitable support material. It is important to note that the substrate molecules and

the products formed should move freely in and out of the phase to which the

enzymes are restrained. Various materials can be used as matrix or support

system for enzyme immobilization, generally which are grouped into three

categories Natural polymers, synthetic polymers and inorganic materials.

Figure 2.20: Covalently immobilization of enzyme with the resin surface (Sirisha, Jain, and Jain, 2016).

Page 79: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

60

Based on support or matrix used for immobilization and type of bond involved in

attachment of enzymes, there are different methods of enzyme immobilization and

various factors impact the immobilized enzymes performance. The different

enzyme immobilization methods are shown in Figure 2.21, and are listed below

(Sirisha, Jain, and Jain, 2016):

• Adsorption / carrier-binding method

• Covalent bonding / cross-linking

• Entrapment method

• Encapsulation / membrane confinement

• Copolymerization

Figure 2.21: Enzyme immobilization methods (Sirisha, Jain, and Jain, 2016).

Cancilla et al. (Cancilla et al., 2000) demonstra ted an other appIication of

immobiIized enzymes, th at is, detect ion of non -active un bound compounds using

ESI-FT-ICR-MS. First, a library of 19 compounds was analyzed by MS and spectra

Page 80: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

61

was obtained as shown in Figure 2.22 A, followed by the incubation of library with

ian iexcess iof iimmobiIized ipepsin enzyme, after that, anaIysis of ithe iunbound

Iibrary members was carried out by MS which results in spectra B in Figure 2.22.

PotentiaI iinhibitors were iidentified by subtraction of the imass spectra ibefore iand

after iincubation. By comparing both spectra, it can be observed that, inhibitors

number 13, 15 and 17 are totally absent in spectra A, which indicates that those

are active inhibitors of targeted enzyme. This technique uses the comparison

method to identify the inhibitors there is no need of enzyme-inhibitor complex

dissociation, which can save an extra step. The MS analysis of compounds must

be performed in both polarities to maximize the ionization efficiency and to ensure

the detection of all compounds. Moreover, the use of high resolution FT-ICR

makes it possible to screen without using an additional HPLC (H. Gao and Leary,

2003; Verdugo et al., 2001).

Figure 2.22: Screening of library of compounds using immobilized enzyme assay, spectra A is before incubation & spectra B is after incubation (H. Gao and Leary, 2003).

Immobilized enzyme technique has various advantages iin icomparison ito

methodoIogies using ienzymes iin soIution, like, easy separation and isolation of

the reactants and products iin ithe enzyme reaction, easy recovery and repeated or

continuous reuse of the enzyme, simultaneous used for both direct and indirect

Page 81: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

62

methods should be mentioned. Immobilization of enzyme is time consuming and

laborious procedure which is a major disadvantage of this method. While

immobilizing the enzyme there is a chance, that active site of enzyme may be

altered or deactivated and changed its characteristics due to steric hindrance.

After every use it becomes more difficult to regenerate the column efficiently,

especially when strong binding inhibitors are infused then, it takes a very long time

to remove then and regenerate the column by buffers. Sometimes, it is impossible

to remove inhibitors without giving damage to the immobilized enzymes (de Boer

et al., 2007).

2.5.5 FrontaI A ffinity Chromato graphy-Ma ss Spectro metry (F AC -

M S)

FrontaI affinity technique uses a different approach to quantify the potential of an

inhibitor toward an enzyme by using the immobilized enzyme technique. In this

technique inhibitors are infused into an enzyme coIumn, whiIe the eIuting inhib itors

or compounds are detecte d b y mass spectrometry, their eluting time and pattern

is used to compare their potential or affinity with a standard (Schriemer, 2004;

Slon-Usakiewicz et al., 2005).

The inhibitors fIowing through the coIumn of immobiIized enzyme are in

continuous equiIibrium with the enzymes. The elution order of compounds

represents their affinity for the target enzyme, with loosely bound compound elute

first and the tightest binding compounds eIuting Iast. In FAC-MS screening

method, a void marker and an indicator are required. Void marker is a compound

which has no affinity for the target enzyme, it will not bind and elute first and has

the shortest elution time, in term of liquid chromatography it is known as solvent

front. The indicator or standard is a compound, whose affinity is known for that

target enzyme, its elution time may be known, or it may be obtained from the initial

experiments. Principle of FAC is shown below in Figure 2.23.

Page 82: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

63

Figure 2.23: Principle of FAC-MS where early eluting compounds has no or low affinity and vice versa (Schriemer, 2004).

Affinity of the other compounds is determined by comparing with the time of elution

of standard with different compounds, a decrease in the retention time of the

standard indicates the presence of strong inhibitors and vice versa. Furthermore,

the MS profile of the indicator gives information about the presence of weak or

strong inhibitors.

Page 83: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

64

Figure 2.24: FAC-MS enzyme inhibitor screening using ‘roll-up height’ phenomenon (Slon-Usakiewicz et al., 2005).

Slon-Usakiewicz et al. introduced a phenomenon of ‘roll-up height’ in enzyme

inhibitor affinity study. If there is an inhibitor in the screening mixture that has

greater affinity for the enzyme target than the affinity of the indicator then this

compound will, before equilibrium is established, displace more of the indicator,

thus generating an over concentration (or peak like shape) of the indicator in the

chromatogram Figure 2.24. By contrast, if the screening mixtures only contain

inhibitors that have weaker affinities for the target than the indicator, then

depending on the degree of affinity, only the indicator will elute earlier (i.e. its front

will shift to the left) and there will be no roll-up (Slon-Usakiewicz et al., 2005).

Simultaneously screening of compounds against two enzymes in a same

immobilized column, is an interesting addition to FAC-MS methodology. In this

method for each enzyme an enzyme-specific indicator has to be added (Chan et

al., 2003). Moreover, F AC-M S offers a appropriate meth od and enables a rapid

ranking and identification of inhibitors in the mixture by measuring the relative

binding strengths or affinity and IC50 values (Slon-Usakiewicz et al., 2004).

Page 84: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

65

2.5.6 Capillary Isoelectric Focusing-Mass Spectrometry (CIEF-

MS)

CIEF-MS is not a very well know techniques for the analysis of enzyme inhibitor

screening, because of its time taking steps and low throughput, In CIEF-MS

inhibitors are incubated offline with the targeted enzyme, followed by pretreatment,

enzyme-inhibitor complexes are separated by CIFE based on their isoelectric

focusing points (Min et al., 2013).

Lyubarskaya et al. used the CIEF- MS for screening against the SH2 domain. In

this method, a pH gradient was formed in the capiIIary and a current was passed

though it. that decreases as the compounds separate and focus at their isoeIectric

points. Afterward, the focused compounds or components of reaction incIuding

unbound compounds, receptor and receptor-Iigand compIexes; were dissociated

and mobiIized by a Iow pressure and finaIIy identified by ESI-MS (Lyubarskaya et

al., 1998). This technique is restricted to the screening for those inhibitors which

have high-affinity toward enzymes, otherwise the enzyme inhibitor compIexes may

be dissociate before analyzing on mass spectrometer.

2.5.7 Flow Injection Analysis-Mass Spectrometry (FIA-MS) and

High-Performance Liquid Chromatography-Mass

Spectrometry (HPLC-MS)

FIA-MS or HPLC-MS analysis for the screening of enzyme inhibitor assays are

most versatile in term of their sample preparation, analysis and output, as well as

these techniques can be enhanced by combining various other steps like used of

column for purification of extracts or compounds prior to incubation with enzymes,

used of robotics and fast chromatography with advance instrumentation to reduce

the assay time etc. In these techniques the reporter molecule, substrate and

product, are mostly used for the monitoring of enzymatic reaction. It comprised to

two steps, incubation of compounds with enzyme followed by the injection of

sample mixture into the MS, as shown in Figure 2.25 (Luque de Castro, 2019;

Norris et al., 2001).

Page 85: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

66

Figure 2.25: FIA-MS in step A preparation and incubation of sample, in step B injection of sample into LC-ESI-MS system (Luque de Castro, 2019).

Screening for the activity of inhibitors is comparatively easy on this system as

demonstrated by Takayama et al. This method was compri sed of two steps,

incubation of reaction mixture comparised of specific compounds or inhibitor,

substrate and glycosidase enzyme, foIIowed by FlA-ESl-MS for dete ction of the

prod uct. This step was repeated for all the compounds and for each injection a

peak cluster was obtained in MS spectra. Spectra of clusters did not provide

information about the inhibition of enzyme, but, if it is converted into the ratio of

product to internal standard, then it gives the reIative quantitation of the product

and by compar ing all th e compounds with standard drug or control, potential of

individual inhibitor can easily be seen.

Page 86: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

67

Figure 2.26: Spectra and results of inhibition study of drugs using FIA-MS (Takayama et al., 1997).

As shown above in Figure 2.26, the 1st and 6th reactions were controled and the

low ratio of compound no. 19 showed that less amount of substrate was converted

to the product, which indicates that compound no. 19 is an inhibitor of target

enzyme. By comparing the ratios of all the individual compounds with the control

the compounds can be ranked by their inhibitory potential (Takayama et al., 1997).

To increase the performance of mass spectrometer and for better quantitation, on-

Iine HPLC can be used before the mass spectrometer; to separate the

componented of enzyme reaction, to avoid the interference of other compounds

on substrate or product. ln 19 97, Wu et al. used the high throughput method

employing HP LC-M S-based scre ening to screen 7 00 compounds in a day. Th is

meth od was abIe to genera te inhibition data by quantifying the reporter molecules

b y means of an Internal standard (Jiangyue Wu et al., 1997).

In 2004, to achieve higher throughput, hig h - speed robotics iand ifast

ichromatography was used by Özbal et al. which led to the fast screening of

compounds (12 sample/min.) against acetyIchoIinesterase (AChE). Higher

throughput couId aIso be iachieved by using muItiple LC systems iinterfaced to a

isingIe mass spectrometer, and iby muItiplexing two ior more isimilar enzymes in ia

singIe reaction imixture (Deng et al., 2004; Özbal et al., 2004).

In LC anaIysis, the throughput is Iimited by the wi dth of chromato graphic peak,

therefore longer run time of LC is not favorable, which can be cope up by using

the modern and fast ultra-performance liquid chromatographs (UPLCs). To g ain

more speci ficity and sensi tivity in detec tion of reporter molecules, selected

Page 87: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

68

reaction monitoring (SRM) or single ion monitoring (SIM) mode can be used. In

this mode MS/MS is per formed ion ithe substrate and iproduct iand itheir specific

ifragments iare used ifor screen ing (Bu et al., 2001; Chu et al., 2000). There have

been many modification and advancement done in the assay monitoring using LC-

MS techniques (Fu et al., 2019; Song, El-Demerdash, and Lee, 2012; C. Wang,

Tian, and Wang, 2011). These methods proved that not only the quantification and

speed of HPLC-MS based screening were reliable, but also accuracy, sensitivity

and specificity of LC-MS method were greater than conventional screening

assays.

2.5.8 HPLC Based Continuous Flow System-Mass Spectrometry

(CFS-MS)

Continuous flow system performed enzymatic assay in an online open-tubular

system which can comprise but not limited to sample injection, HPLC separation,

an on-line biochemical assay and mass spectrometric detection. In CFS

compounds are continuously added and mixed with the enzymatic assay reagents,

substrate, buffer etc., resulting in continuous output of data, generated in the form

of spectra by MS that refIects the enzymatic activity. Potential inhibitors separated

by HPLC can bind to the enzyme and reduce its activity, ultimately decrease the

product formation, which results in a decrease of product signal. MS data from the

inhibitor is obtained simultaneously.

Most CFS-MS-based screening methods used fIuorescence detection and

fIuorescence-labeled substrate to detect active compounds. Those systems split

the HPLC effIuent to the enzymatic assay and to the mass spectrometer, to

provide information about the inhibitors and by comparing the retention times of

activity peaks of both detectors led to identification of the inhibitor. Recent

advancement in MS instrumentation and speed of assay make it possible to use

non-labeled substrate and drugs and detect them both via mass spectrometer at

both in effluent and split volume of initial injection. A schematic diagram of CFS-

MS is given in Figure 2.27 (Ingkaninan, de Best, et al., 2000; Ingkaninan,

Hazekamp, et al., 2000; Rhee et al., 2004; van Elswijk et al., 2003). In CFS,

substrate is continuously infused through the enzyme-reactor column (immobilized

Page 88: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

69

enzyme column). In the absence of inhibitor, the infusion of substrate led to the

formation of a certain amount of product. Enzyme inhibition was detected by

changes in the substrate-to-product ratio (Hodgson et al., 2005).

Figure 2.27: Continuous flow system with MS detection using sample injection and online reaction of enzyme and substrate in the presence of inhibitors (Ingkaninan, Hazekamp, et al., 2000).

On-line coupling of HPLC with MS and biochemical detection proved to be

advantageous over the traditional fractionation approaches in terms of

dereplication speed and resources required, and for isolation of active

compound(s) from crude extracts. By using this technique all the components of

the enzymatic reaction can be detected as well as it can also be used for the

multiplex assay.

2.5.9 Matrix Assisted Laser Desorption Ionization-Mass

Spectrometry (MALDI-MS)

MALDI-MS is a widely applied tool for the analysis of proteins and peptides

because of its relatively high tolerance against salts and biological contaminants

as compared to the ESI-MS, as well as its excellent detection limits, speed of

sample analysis, and its ease of data analysis make it suitable for proteomics and

peptidomics (de Boer et al., 2007).

The emergence of the first MALDI based report dedicated to screening

applications was published in 1998 by (Hsieh, Keshishian, and Muir, 1998). This

Page 89: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

70

group reported the development of automation (via liquid handling robotics) to

prepare and spot up to 4,000 reaction mixtures on a single 2x2 inch target plate.

Importantly, they showed the potential for multiplexing to evaluate inhibition of

several enzymes simultaneously. Unfortunately, they did not address quantitative

issues and simply reported the results as a qualitative profile of whether a

compound inhibited the enzyme or not. Nonetheless, this work demonstrated the

feasibility of using the rapid and sensitive acquisition of MALDI-TOF for measuring

enzyme activity for screening inhibitory compounds.

In 2006, Greis et. al. published a report, in which they demonstrate how MALDI-

TOF MS can be used to quantify the inhibitory potential of drugs in term of their

IC50 values. In this report ratio of substrate and product was used to produce IC50

curves for rapid enzyme assays and compound screening. Typical reproducibility

parameters were less RSD as compared to other techniques and quantitative

assays and well within the acceptable limits for screening assays. The speed of

the MALDI readout is currently about 10 s per sample, thus allowing for over 7500

samples per day, while enzymatic reaction mixtures are prepared by liquid

handling robots. Importantly, the ratios of substrate to product are of enough

reproducibility to eliminate the need for internal standards and, thus, minimize the

cost and increasing the speed of assay development (de Boer et al., 2007; Greis

et al., 2006; Park et al., 2012).

The rapid ionization and desorption features of advanced matrix-assisted laser

desorption ionization-triple quadrupole (MALDI-QqQ) mass spectrometer are

shown to improve the speed of enzymatic analysis to greater than 1 sample per

second (Rathore et al., 2008).

Most recent publication for the quantitation via MALDI-MS showed that,

temperature-selected MALDI spectra had greater reproducibility, which can be

changed intο the direct propοrtiοnality between the analyte-tο-matrix iοn

abundance ratiο and the analyte-tο-matrix ratiο in the sample, allοwing easy

quantitation οf the analyte. The relatiοn has been fοund tο hοld, even when the

analyte is a cοmpοnent οf a mixture. Another advantage of the methοd is that, οne

can quantify an analyte withοut adding an internal standard. The method will

becοme an inexpensive technique suitable fοr quick quantitative screening οf any

Page 90: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

71

analyte analyzable on MALDI such as proteins and peptides. The fact that the

cοncentration of analyte is prοportional tο the analyte-tο-matrix iοn abundance

ratiο can be used fοr quick cοmparison of the relative amοunts οf a particular

analyte in twο οr mοre biοlogical samples (Bae, Park, and Kim, 2012; Park et al.,

2012).

2.5.9.1 Multiplex Analysis

One of the most important feature of enzymatic assay analysis on MALDI-MS is

the easiest multiplex assay. In multiplex assay one compound or drug can be

screen against two or more enzyme at a time if each enzy has different substrate

and product.

In an example by Hsieh in 1998, a multiplex assay was developed using three

enzymes; angiotensin converting enzyme (ACE), protein tyrosine phosphatase

(PTPase) and N-myristoyltransferase (NMT), which used the substrates

angiotensin I, PTPase substrate TRDIXETDYYRK, where X = Y-PO3H2, 10

pmol/L) and NMT substrate GNAASARR-NH2 with myristoyl=CoA, respectively

(Hsieh, Keshishian, and Muir, 1998).

Page 91: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

72

Figure 2.28: Multiplex assay on MALDI-MS (A) is the spectra of cοntrοl withοut any inhibitοr, (B) is spectra of with the inhibitοr Lisinοpril (Hsieh, Keshishian, and Muir, 1998).

Page 92: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

73

Chapter 3: Method Development for Screening

of ACE inhibitors Using HPLC-ESI-QqQ-MS

Page 93: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

74

3.1 Introduction

Several methods involving the measurement of ACE activity and inhibition activity

of drugs have been described, including spectrophotometric (Holmquist, Bünning,

and Riordan, 1979; Li et al., 2005), biochemical (van Elswijk et al., 2003),

fIuorimetric (Cheung et al., 1980; Kang et al., 2002), high-performance liquid

chromatography (HPLC) (Chen et al., 2013; Lahogue et al., 2010; Jianping Wu,

Aluko, and Muir, 2002), internally quenched fIuorogenic (Araujo et al., 1999) and

mass spectrometric methods (Greis et al., 2006; Hsieh, Keshishian, and Muir,

1998; Xiao et al., 2006). Most of the previously mass spectrometric method used

the LC-MS or MALDI-TOF but this developed method used the LC-MS/MS

approach with very low level of quantification to monitor the enzymatic reaction. A

comparison of this method with previously developed methods is also given in the

Table 3.9 at the end of this chapter.

High performance Iiquid chromatography-mass spectrometry (HPLC-MS) is a very

highly sensitive and selective technique, which is a very useful tool for the field of

biological analysis and it is extensively used for the identification and quantification

of peptides and proteins. When LC-MS combines with the MS/MS then it becomes

a very unambiguous and sensitive analysis techniques for peptides in compIex

sampIe matrices (de Boer et al., 2007; de Rond, Danielewicz, and Northen, 2015;

Greis, 2007; Liesener and Karst, 2005; Rathore et al., 2008).

During this study a HPLC-ESI-MS/MS based imethod iwas deveIoped ifor

simuItaneous anaIysis iand iquantification iof iangiotensin l in ian ienzymatic imixture

by iusing muItipIe ireaction imonitoring (MRM) imode. The analyses iconditions for

iHPLC separation iand iMS idetections were ioptimized iusing a istandard imixture of

angiotensin I, angiotensin II and Bradykinin (internal standard). The imethod iwas

evaluated ifor reproducibility, precision, Iimit of quantification and idetection. Two

commercially available antihypertensive drugs, Captopril and Lisinopril, were

checked to validate the method. The IC50 values of both standard drugs (Captopril

and Lisinopril) were determined and used as reference for the screening of other

inhibitors (drugs and compounds). The newly developed assay offers the

possibility to use only 20 pico-mole amount of substrate (angiotensin I) per well,

Page 94: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

75

as this low quantity was found to be sufficient to perform a complete time

dependent study and to determination the IC50 value. This in turn also decreased

the amount of inhibitors (drugs and compounds) used in inhibition assay, up-to

pico-mole level quantity.

3.2 ExperimentaI

3.2.1 Chemicals and Reagents

Angiotensin converting enzyme, its substrate angiotensin I, product angiotensin II,

and internal standard bradykinin all were purchased from Sigma AIdrich (USA).

Standard inhibitors Captopril iand Lisinopril were purchased from Tokyo Chemical

Industries Co, Ltd. (Japan), both were >98% pure. Tris buffer (research grade)

was purchased from Serva (Germany). The concentration of this buffer was

prepared 20 mM with 3 mM dithiothreitol (DTT) for the pH 7.5 at 37 oC. Formic

iacid iand acetonitriIe iwere ipurchased from iFisher iScientific (Leicestershire, iUK).

All solvents used were of spectroscopic grade, fresh MiIIi-Q water was used from

MiIIi-Q water assembIy (Bedford, iUSA) which had 16.5 MΩ resistance.

3.2.2 Calibration Curve

To plot the calibration curve for angiotensin I, a concentration range of 20-200 nM

with 100 nM of internal standard (Bradykinin) were prepared from the bulk

concentration, in 0.1% formic acid solvent. For the calibration curve, 5 different

initial concentrations of 100, 300, 500, 700, and 1000 nM were diluted up to 5

times with 0.1% FA and the internal standard bradykinin. The final concentrations

of calibrators were 20, 60, 100, 140, and 200 nM with 100 nM of internal standard.

A scheme of sample preparation is given in Figure 3.1. The calibration curve was

prepared by pIotting the ipeak iarea iratio iof anaIyte-to-internaI istandard ito the

iconcentration of anaIyte.

Page 95: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

76

Figure 3.1: Scheme for samples of calibration curve.

3.2.3 Angiotensin Converting Enzyme Assay

Enzymatic iassay iwas iperformed iin ia i96-weII pIate with ia totaI voIume of i25 iμL ifor

ieach ireaction. Each weII contained i2.5 iμL iof iangiotensin converting ienzyme

(ACE) i(0.2 μM), 20 iμL iof isubstrate angiotensin l (1 iμM) iand i2.5 iμL iof istandard

inhibitors iof idifferent iconcentrations iin different weIIs. ln ia controI weII, 2.5 iμL of

iTris ibuffer iwas iadded iaIong iwith ithe ienzyme iand isubstrate to compensate ifor ithe

voIume of ithe iinhibitor. iAssays iwere performed iin ithe incubator iat 37 oC. iBoth ACE

iand substrate iwere iprepared in iTris ibuffer iof i20 imM with i3 imM DDT iat ipH i7.5.

3.2.4 Sample Preparation for LC-ESI-QqQ-MS Analysis

Aliquots of 2.5 μL were ta ken a t differ ent tim e interva ls (5 , 1 0, 1 5, 2 0, 3 0, 4 0, 5 0,

and 6 0 minutes) and mixed with 2.5 μL of 0.5 μM of bradykinin (Internal standard)

and 7.5 μL of 0 . 1 % f ormic acid soIution. F ormic acid soIution (0 . 1 %) was used to

quench the reaction and to make the 1/5th dilution of the reaction mixture. The final

concentration of the internal standard was 100 nM. The initial concentration of

Angiotensin I was 160 nM after dilution. From this mixture duplicate runs were

carried out for the analysis. In each run, 5 μ L o f t he sample was inje cted in to th e

ESI-QqQ- M S sys tem of AgiIent equipped with HPLC system and ESI Jet Stream

Source. Agilent Mass Hunter Data Acquisition 5.0 and Data Analysis 6.01 were

used for data acquisition and interpretation, respectively.

Page 96: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

77

3.2.5 LC-ESI-QqQ-MS AnaIysis

AII isampIes iwere anaIyzed usi ng AgiIent 12 60 Iiquid chromatograp h (AgiIent

TechnoIogies, WiImington, DE ), coupIed with AgiIent 64 00 tripIe quadrupIe mass

spectrometer (AgiIent TechnoIogies, WiImington, D E, U SA). S eparation o f

peptides was a chieved using reverse ph ase HPLC with Jupiter C-18 column of

Phenomenex (Torrance, California) (50 mm length, 1 m m i.d ., 5 µ m parti cle si ze)

a t 25 oC. The mob ile ph ase contained water with 1% formic ac id as soIvent A, a nd

acetonitriIe wit h 1 % f ormic a cid as soIvent B. A gradien t of 5-70% of B was run f or

a totaI tim e of 7 minutes. T he gradient w as started with 5% B, which was changed

progressively to 70% B in 3 minutes, and then rapidly decreased again to 5% B in

half minute and run for the next 4 minutes at same composition. The fIow r ate w as

optimized a t 0 . 4 m L/ min . a scheme of gradient flow is given below in Figure 3.2.

Figure 3.2: Scheme of solvent gradient on HPLC.

The Agilent Jet stream source was optimized for all peptides to give the maximum

response (peak height) and S/N value. Among the 7 parameters, 5 were optimized

by using a statistical software “Design Expert V9.0”. To optimize and verify the

source parameters and fragment chosen, a software of Agilent “MassHunter

Optimizer Version B.01.00” was also used for the optimization of peptized, which

Page 97: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

78

run independently and give the final results. The final values were selected as

follows: gas temperature 300 oC, gas fIow 12 L / min, nebuIizer pressure 30 p s i,

sheat h gas heate r 240 oC, shea th ga s fIow 10 L / min, capiIIary voItage 30 00 V an d

nozzIe voItage ( V Charging) 1000 V. MuItipIe re action moni toring (M R M) mode

w as used to coIIect ma ss spectraI data of pr ecursor and product ion tr ansitions.

T he coIIision ene rgy a nd th e f ragmentor voItages were t uned wi th respec t to

individuaI anaIyte to maxi mize the anaIyte respon se, as gi ven beIow in Table 3.1

Table 3.1: MRM transitions and source conditions for fragmentation.

Type Name Precursor

I on (m / z)

Product

I on (m / z)

Dw ell

Time

(ns)

Fragmentor

Voltage ( V )

Collision

Energy

(e V)

Ce ll

Acc

Internal

Standard Bradykinin 530.8 175.2 60 100 30 7

Substrate Angiotensin I 433 110.1 60 100 20 7

Product Angiotensin II 349.7 136 60 80 10 7

3.3 Results and Discussion

The data for the three peptides including angiotensin I (substrate), angiotensin II

(product) and bradykinin (internal standard) was acquired using optimized LC-

MS/MS conditions. The ratio of analyte-to-internal standard only for angiotensin I

was used for further calculation, calibration curve, and enzymatic analysis.

3.3.1 Optimization of LC-MS/MS Analysis Conditions

Separation efficiency o f the peptides was checked using two Jupiter C18 columns

(Phenomenex Inc., Torrance, California.) of different dimensions, column-1 of 1

m m i .d., 5 cm length with particIe si ze o f 5 μm, and column-2 of 0.5 mm i. d., 1 5

c m Iength with particIe si ze o f 5 μm. Column-1 was further used for separation.

Page 98: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

79

Column-2 was found t o b e not suitabIe f or the hig h - throughpu t anaIysis because

o f very low optimum flow rate, which significantly increased the run time for

sufficient separation of peptides. Column-1 was tried with different gradients and

different flow rates, and optimum was found to be 0.4 mL/min with gradient of 5-

70% B in 3 minutes. Peaks for the peptides of substrate and product were eluted

within 5 minutes and showed sufficient separation for analysis, as shown in below

spectra Figure 3.3.

Figure 3.3: Total ion chromatogram of peptides.

For angiotensin I & angiotensin II, the parent ion of +3 charges was selected while

for bradykinin the parent ion of +2 charges was selected as precursor ions due to

their higher intensities. Collison energy and other mass spectrometric parameters

were optimized for each peptide are summarized in Table 3.1. Fragments

(quantifier) of highest intensity were selected and optimized for MRM transition as

m/z 530.8→175.2 for bradykinin, m/z 433→110.1 for angiotensin I and m/z

349.7→136 for angiotensin II. Maximum intensities for these fragments were

achieved at collision energy values of 30, 20, and 10 eV for angiotensin I,

angiotensin II, and bradykinin (IS), respectively. Retention time and MRM spectra

of each standard can be seen in Figure 3.4.

Page 99: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

80

Figure 3.4: Retention time and MRM transition of bradykinin, angiotensin II and angiotensin I, in a standard mixture.

3.3.2 Optimization of Jet Stream Source Using Design Expert

3.3.2.1 Optimization of Ion Source

Design iExpert iis ia statistical isoftware ipackage ifrom iStat-Ease iInc. which iis

specificaIIy dedicated itο iperforming the idesign iοf iexperiments (D OE). iDesign

iExpert ioffers icomparative itests, icharacterization, iοoptimization, irοbust iparameter

idesign, imixture idesigns and combined idesigns. Design iExpert prοvides itest

matrices ifοr up itο i5 0 ifactοrs. GraphicaI tοοIs may heIp tο identify ithe iimpact οf

each ifactοr iοn the idesired οutcοmes iand can give ithe οptimize vaIue fοr a specific

functiοn (Comley, 2009; Hooda et al., 2012). In this work design-expert was used

to optimize the parameters of ion source of ESI-QqQ-MS,

Methοd deveIοpment can be perfοrmed by appIying a οne variabIe at a time

apprοach, where a singIe parameter is varied, whiIe aIl the οthers are kept

cοnstant. Hοwever, this apprοach is οften insufficient fοr cοmprehensive studies

Page 100: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

81

since it dοes nοt cοnsider the interactiοns amοng the individuaI parameters. A

mοre carefuI apprοach can be adapted by making an experimentaI design which

emphasizes the reIatiοnships amοng the variοus parameters and requires a Iοwer

number οf anaIyses (Greco, Boltner, and Letzel, 2014).

For ESI Jet Stream source, five out of seven parameters were optimized by using

the software. G as te mperature, ga s fIow, nebuIizer pres sure, she ath g as fIow and

capiIIary voItage we re op timized by software while sheath gas temperature and

nozzle voltage were adjusted manually. Different range of each parameter was

provided to the software as shown in Table 3.2 and number of experiments were

limited to nineteen (19) including a mid value experiment and one random

experiment were also be run along with the designed experiments, the scheme

was as follows in Table 3.3.

Table 3.2: Range of different parameters feed to the software.

Parameter (Unit) Range

Gas temperature (oC) 120-300

Ga s fIow (L/min) 3-12

Neb ulizer pre ssure (p s i) 30-60

Shea th ga s fIow (L / min) 6-10

Capil lary vοl tage (V) 3 0 0 0-5500

Page 101: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

82

Table 3.3: Experiments design output of Design-Expert.

Experiment

No

Gas

temperature

(oC)

Gas fIow

(L / min)

NebuIizer

pres sure

(p s i)

She ath

g as fIow

(L / min)

CapiIIary

vοItag e

( V)

1 300 3 60 6 5500

2 300 12 60 10 5500

3 300 3 30 6 3000

4 120 12 60 10 3000

5 300 3 30 10 5500

6 120 12 60 6 5500

7 300 12 60 6 3000

8 120 3 60 6 3000

9 300 12 30 6 5500

10 300 12 30 10 3000

11 120 3 60 10 5500

12 300 3 60 10 3000

13 120 3 30 6 5500

14 120 12 30 10 5500

15 120 3 30 10 3000

16 120 12 30 6 3000

17 210 7.5 45 8 4250

18 255 7.5 52.5 9 4875

19 165 7.5 37.5 7 3625

20

(random

values)

200 10 50 5 3500

Page 102: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

83

Table 3.4: Response of bradykinin and angiotensin I correspond to each experiment.

Experiment No Response 1:

Bradykinin (peak area)

Response 2:

Angiotensin I (peak

area)

1 277 546

2 548 568

3 748 803

4 439 822

5 400 663

6 214 307

7 844 861

8 128 267

9 762 897

10 1541 1699

11 237 645

12 150 465

13 795 1286

14 400 991

15 200 571

16 286 646

17 438 647

18 314 326

19 289 408

20 277 546

Page 103: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

84

Figure 3.5: Response of bradykinin and angiotensin I.

Data for all experiments was tabulated in software as given in Table 3.4, and

converted into the graphical representation as in Figure 3.5. From the data it was

found out that, the most suitable conditions for the both peptides were at extreme

conditions of parameters ranges, in the experiment number 10, therefore

parameter values of experiment 10 were selected and used for further

reproducibility.

As in ESI peptides ions can be produced with single, double and triple charge, to

find out that which of the species is produced in high intensity, another series of

experiment was performed similar to the above, as given in Table 3.5, to check

the intensity of singly, doubly and triply charged ions by using the 5 experiments

including the experiment 10 conditions.

Page 104: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

85

Table 3.5: Checking of ionization of multiple charge ions of angiotensin I and bradykinin.

Experiment No

Angiotensin I Bradykinin

Single Charge

1297.5 Da

Double Charge

648.8 Da

Triple charge

433.4 Da

Single Charge

1060.3 Da

Double Charge

530.7 Da

Triple charge

354.3 Da

3 7129 107208 104053 17448 402278 64101

7 5482 125038 136398 18067 506201 67569

10 6704 177766 215042 16602 741156 32712

13 5687 134300 182482 13309 627838 67234

20 1596 100950 108685 3942 342539 22675

Page 105: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

86

Figure 3.6: Comparison of multiple charge ions of angiotensin I (above) and bradykinin (below).

Data of peak area of each ion was converted into graphical form as given in Figure

3.6. It was observed that, in angiotensin I, in all experiments the intensity of triply

charged (3+) ions is very high, intensity of doubly charged (2+) is nearly

comparable to 3+ ions but it is less than 2+, while the intensity of singly charged

(1+) ions is very low. In bradykinin the doubly charged (2+) ions has highest

intensity while singly and doubly charged (1+ & 2+) ions has very low intensity.

The angiotensin I is a decapeptide therefore it can accommodate triple charge,

Page 106: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

87

while bradykinin is an octapeptide therefore it can have double charge while triple

charge is rare in bradykinin, therefore for analysis angiotensin (3+) and bradykinin

(2+) was used.

3.3.3 Mass Hunter Optimizer for Peptides

Along with Mass hunter data acquisition and analyzer Agilent has provided a

software “MassHunter optimizer for peptides Version B.01.00”, to optimize the

source parameter as well as MS/MS (MRM) conditions for compounds and

peptides.

This software requires 4 steps to start auto optimized any compound or peptides.

In step 1 (Figure 3.7 A) mode of injection of sample, fragmentor range and

collision energy range is needed to be set, for optimization of Angiotensin I

“automatic injection using loop injection” was used, fragmentor range was selected

from 60-140 and collision energy range of 5-40 was used. In step 2 (Figure 3.7 B)

mode of analysis and charge state of ions was selected, for AI positive mode with

charge state 1,2 & 3 was selected. In step 3 (Figure 3.7 C) lower limit of mass of

ions or fragments was selected, for AI lower limit was set to 80, to avoid

suppression of other fragments. In last step (Figure 3.7 D) Sequence of peptide

was needed to be entered in single alphabet format along with mass and sample

position of peptide in auto sampler.

Optimizer worked in a sequence and performed a number of runs by changing

some parameters in each run, and then optimized the parameters one by one, in

the end it provides the results in excel file, with has values of source parameters

and best fragment along with MRM conditions. The results of these software were

almost same as obtained by previous optimization, except gas flow and

temperature, which were little bit higher, but when manually optimized, previous

parameters were gave the better results. Optimizer also choose the 110 m/z

fragment of AI as best fragment along with same MRM conditions, which further

confirms the manual optimization of AI. Steps of MassHunter Optimizer for

Peptides are shown in Figure 3.7.

Page 107: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

88

Figure 3.7: Steps of ‘MassHunter Optimizer for Peptides’.

Page 108: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

89

3.3.4 Determination of Calibration Curve, LOD and LOQ

Concentration range for angiotensin I calibration curve was chosen as 20-200 nM,

because the initial concentration of angiotensin I in the enzymatic reaction was

160 nM. For the calibration curve, 5 different calibrators of 20, 60, 100, 140, and

200 nM of angiotensin I were prepared with 100 nM of internaI sta ndard

bradykinin. The caIibration cu rve s howed a linear response ov er th e selected

ra nge o f concentrations (y=0.0888x-0.1932) with linear regression of R2=0.999,

Figure 3.8.

Figure 3.8: Calibration curve of angiotensin I.

The Iimit of d etection and Iimit o f quantification f or th e angiotensin I was caIcuIated

usi ng t he standar d dev iation o f th e res ponse ( σ ) (replica of different concentrations

used in caIibration cur ve) and t he sIope (m) of caIibration curve i .e . L O D = 3 σ / m

an d L O Q = 1 0 σ / m. LO D a nd L OQ we re fo und t o b e 1.44 nM ( 1.866 n g /m L) an d

4.37 nM (5.664 n g /m L), respect iveIy.

Fo r intra day an d inter day precis ion (RSD, %) an d ac curacy (% error) o f calibration

curve, two QC samples of concentrations 80 and 180 nM were analyzed. For

intraday analysis three samples of each concentration were analyzed in triplicates

Page 109: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

90

in a day and for interday one sample of each concentration was analyzed in

triplicate on three different days in a week (day 1, day 4 and day 7). The iaccuracy

iwas caIculated from ithe iexpected iconcentration (C E) and ithe imean ivalue iof

measured iconcentration (C M) iby iusing ithe foIIowing reIation:

Accu racy (% error) = C � − C � C�

× 1 0 0 (. 3.1)

SimiIarly, the irelative istandard ideviation, iRSD iwas determined ias ia measure iof

iprecision from ithe istandard deviation iand the imean ivalue of the imeasured

concentration iby empIoying ithe foIIowing relation:

Precisi on "R S D %& = St andard dev iation (S D)C �

× 10 0 (. 3.2)

For both qualifiers, RSD% was in the range of 1.76 - 4.37, results of intraday and

interday are summarized below in Table 3.6.

Table 3.6: Intraday and Interday accuracy and precision determination of angiotensin I.

Angiotensin I

Conc. (nM)

Intraday Interday

Mean

Conc. (nM)

Precision

RSD % %Error

Mean

Conc. (nM)

Precision

RSD % %Error

1. 80 80.31 4.33 -0.38 77.52 4.37 3.09

2. 180 176.69 1.76 1.84 184.05 3.27 -2.25

3.3.5 Enzymatic Reaction

For the enzymatic reaction, angiotensin I was used as a substrate because of its

low Km (Michaelis-Menten constant) value (El-Dorry et al., 1982; Sakharov,

Danilov, and Dukhanina, 1987) as well as it has low LOD/LOQ and linear range

as compare to other substrates, as shown in the comparison table in the section

“Comparison with Reported Methods”. The enzymatic assay was comprised of

Page 110: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

91

total reaction volume of 25 μL, which contain 20 μL of substrate (800 nM). For

analysis, 2.5 μL of the reaction mixture was taken at different time intervals and

diluted 5 times with 1% formic acid and internal standard, which makes the

concentration of substrate equal or less than 160 nM, while injecting into the MS

instrument. The enzyme activity was monitored by direct quantification of the

consumption of substrate by the equation 3.

%C = )1 − *+, × 100 (. 3.3)

Where %C is the percentage conversion, S is the initiaI concentration of substrate

and X iis ithe iconcentration iof ithe isubstrate after a specific time. The concentration

of the enzyme, incubation time, and temperature were optimized at constant

concentration of substrate. The enzyme activity was checked at 25 oC and 37 oC

from which it was inferred that the enzyme is more active at 37 oC, therefore used

for further assay.

Enzymatic reaction can be monitored by two ways, either by increasing

concentration of product or by decreasing concentration of substrate. If the assay

is monitored by the product concentration, then the Iimitation is to monitor the

reaction at a time where more than 30-40% of the substrate is converted into the

product to get a good signal-to-noise ratio. However, if the assay is monitored by

substrate concentration, then an assay can be optimized at a condition where the

product never exceeds more than 10-20% of substrate concentration, which then

ultimately gives a high signaI - t o - noise iratio iand ireduces ithe time of analysis. For

example, at 20% conversion the theoretical concentrations of substrate

(angiotensin I) and product (angiotensin II) will be 128 and 32 nM, respectively,

therefore in both of these, substrate (angiotensin I) can be determine with good

sensitivity.

To idetermine ithe iconditions iat iwhich ithe enzyme reaction imaintained ia iclose

approximation ito ithe initiaI rate, three different enzyme concentrations 0.2, 0.5,

and 0.8 µM were evaluated through a time course of 60 minutes as shown below

in Figure 3.9.

Page 111: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

92

Figure 3.9: ACE assay with different concentrations of enzyme and time course.

The incubation time and enzyme concentration were chosen, so that in initial time

(10-20 minutes) 20-30% of the substrate converted into the product, as this

percentage of conversion will give an approximation of the initial velocity.

3.3.6 Initial Velocity of Enzyme

With the direct quantification of substrate, it is possible to calculate the initiaI

veIocity iof ithe ienzyme reaction which ican ibe used for ithe kinetics study iof that

particular enzyme. In the enzymatic reaction, the concentration of the substrate

(AI) decreases rapidly in initial stages, that is when enzyme has high velocity, but

after some time the velocity of the enzyme decreases gradually. This can be seen

in Figure 3.9, in initial time (0 to 20 min.) percentage conversion increase rapidly

when enzyme had high velocity, but after 20 min it increase slowly when enzyme

had low velocity. The initial velocity of the enzymatic reaction can be calculated

from the few initial points of percentage conversion versus time course graph

(Figure 3.9). ACE velocity was calculated at four initial points of time 5, 10, 15,

and 20 minutes. These four initial points were selected because of their linear

trend, and velocity was calculated by using the following formula:

Page 112: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

93

v� = C- − C.t- − t. (eq. 3.4)

Where vo is the initial velocity of enzyme, Cf is the concentration of angiotensin I

at time tf (f=final), and Ci is the concentration of angiotensin I at time ti (i=initial).

For this ACE assay, the initial velocity of enzyme was found to be 10.385 nM/min,

calculation of initial velocity are given in Table 3.7.

Table 3.7: Initial velocity of ACE in enzymatic assay.

Points Cf – Ci (nM) tf – ti (min.) vo (nM/min)

1 12.278 3 4.093

2 72.824 5 14.565

3 55.952 5 11.190

4 58.453 5 11.691

Average vo 10.385

3.3.7 Determination of IC50 of Inhibitors

Once the enzyme assay parameters and reproducibility were established,

standard drugs or compounds can be investigated for their inhibitory potential. iFor

iconcentration idependent iinhibition istudies, the iinhibition iwas iplotted ias

%inhibitory iactivity (% I A). iTo imeasure the iinhibition iof the ienzyme iactivity, the

idegree to iwhich ithat iactivity i(% C) is icurtailed was measured. iThus, ithe

iconcentration idependent iinhibition idata was pIotted ias %IA versus log of

concentration of inhibitor, iwhere ithe inhibitory iactivity iis the %conversion

imeasured iin controI reactions (without iinhibitor) divided by ithe %conversion

measured in ia reaction with inhibitor, as represented in equation (Chen et al.,

2013; Lahogue et al., 2010):

Page 113: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

94

%Inhibition activity = )1 − %conversion with inhibitor%conversion without inhibition, × 100 (eq. 3.5)

The % inhibition activity is defined as the measurement of the degree of inhibition

and it is used to determine the IC50 value.

Two standard inhibitors of ACE, Lisinopril and Captopril, were checked for the

authentication of the assay. All reaction mixtures and blank assays were

performed and analyzed by using newly developed method. For the determination

of %IA and IC50 value, ia igraph iof i%inhibition activity iversus ilog iof different

concentrations iof iinhibitor iwas used. The point where %IA is 50% on y-axis, there

concentration of inhibitor will be the IC50 on x-axis, as shown below in Figure 3.10.

Figure 3.10: Inhibition of ACE by Captopril and Lisinopril.

For Captopril concentrations of 1, 5, 10 and 50 μM and for Lisinopril concentrations

of 0.5, 1, 5 and 10 μM were used. iAs ithe iconcentration iof CaptopriI or Lisinopril

iincreased, ithe conversion iof ithe substrate to product idecreased. At higher

concentrations of both inhibitors, no significant conversion of substrate into

product was observed. From the graphs, ithe iIC50 values of Captopril iand Lisinopril

iwere ifound ito ibe 3.969 μ M a n d 0.852 μ M, re spe ctiveIy.

Page 114: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

95

3.3.8 Comparison of Captopril and Lisinopril Inhibitory Potential

Determination of IC50 value is the initial step of drug discovery process. A drug can

only be useful if it can demonstrate selectivity for the specific enzyme and has

inhibitory potential, comparable to the standard inhibitors. Inhibitory potential of

inhibitors can be compared by their IC50 values. For comparison of different

compounds or standard a graph between %IA versus log of concentration can be

plotted. In comparison of these two standards inhibitors, Lisinopril was found to be

more potent than the Captopril which is in agreement with the reported data

(Hsieh, Keshishian, and Muir, 1998).

However; IC50 value of an inhibitor may not be exactly same if it is determined by

two different methods, because the IC50 value is highly method depended, it

depends upon the number of factors, like: type of enzyme used (from porcine

kidney, rabbit lung, etc.) because of their activity; concentration and amount of

enzyme; amount, type and concentration of substrate used (A1, HHL, etc.);

incubation time, buffer conditions etc. (Cer et al., 2009). The IC50 value for the

synthetic inhibitors is usually in the nanomolar range and for angiotensin I it is in

the micromolar range (Henda et al., 2013).

3.3.9 Qualification of ACE Assay

Developed method for angiotensin converting enzyme (ACE) inhibition underwent

basic qualification tests to assess its applicability to a screening activity; a full

validation was not considered necessary for the scope of this work. For intraday,

three enzymatic reactions were carried out for each parameter (1. without inhibitor,

2. with Captopril 5µM and 3. with Lisinopril 5µM) and each reaction was analyzed

in triplicates. For interday one enzymatic reaction was carried out for each

parameter on three different days in a week (day 1, day 4 and day 7) and also

analyzed in triplicates. The percentage conversion was averaged from triplicates.

Intraday analysis showed RSD% of 4.14 for the ACE assay without inhibitor, while

for the interday analysis RSD% was 7.42. The ACE assay with inhibitors shows

4.47 and 7.94 RSD% for Captopril, and 9.48 and 9.98 RSD% for Lisinopril intraday

and interday analysis, respectively, as shown below in Table 3.8.

Page 115: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

96

Table 3.8: Intraday and interday precision and reproducibility of ACE inhibition by Captopril and Lisinopril.

ACE

Assay

Intraday Interday

ACE

%conversion

Precision

RSD %

% Error ACE

%conversion

Precision

RSD %

%

Error

1. Without

inhibitor

24.67 4.14 0.94 25.87 7.42 -3.88

2. Captopril

5 µM

12.80 4.47 -10.92 12.07 9.48 -4.56

3. Lisinopril

5 µM

3.48 7.94 1.32 3.34 9.98 5.38

3.3.10 Comparison with Reported Methods

The developed method has very Iow Iimit of quantifi cation a nd detection of

substrate, i n compari son with previously reported methods of ACE. iThis imethod

iprovides exceIIent isensitivity iwithin aIIowable Iimits iof istandard ideviation iand

ierror. iMoreover, the amount of substrate and enzyme needed in the assay is also

very low which can reduce the running cost of assay by many folds. A icomparison

iof ithe deveIoped imethod iwith iother ireported imethods is provide below in Table

3.9.

Page 116: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

97

Table 3.9: Comparison of the current developed method with previously reported methods.

Me th od /

Technique

Line ar ran ge

(µM)

LOD /

LOQ

Substrate Amount of

enzyme used

(per well)

Amount of

substrate used

(nmol /well)

RSD % Ref.

HPLC-ESI-

MS/MS

0.02-0.20 1.44nM

4.37 nM

AI 1.25 pmol

(0.00319 mU)

0.02 ≤9.98 (Musharraf et al.,

2017)

Current method

HPLC-DAD 0.10-1.0 NA HHL 20 nU 108.5 (Jianping Wu,

Aluko, and Muir,

2002)

HPLC NA NA HHL

FA-PGG

4.6875 mU

0.0025 mU

450

262.5

≤18

≤12

(Shalaby, Zakora,

and Otte, 2006)

HPLC NA NA HHL

FA-PGG

1 mU

2.5 mU

500

0.25

(Lahogue et al.,

2010)

HPLC 7.28-465.69 NA HHL 1 mU 75 ≤1.11 (Chen et al., 2013)

VSP 165.32-

1164.23

NA HHL 1 mU 75 ≤5.53 (Chen et al., 2013)

Page 117: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

98

HPLC-ESI-MS 0.446-446.5

(HA)

13.97 nM

20ng/mL

HHL

HGG

NA 200 ≤11.18 (Xiao et al., 2006)

MALDI-TOF-

MS

NA NA AI 1.8 pmol 0.08 NA (Hsieh, Keshishian,

and Muir, 1998)

AI = Angiotensin I, HA: Hippuric acid, HLL: N-Hippuryl-His-Leu hydrate, HGG: Hippuryl-glycyl-glycine, FA-PGG: N -[3 -(2-

FuryI)acryIoyI]-Phe-GIy-GIy

NA: Not available

Page 118: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

99

3.4 Conclusion

LC-ESI-QqQ-M S ioffers ia vaIid readout ifor ienzyme iinhibition screening iassays.

The ispeed, isensitivity, ireduced icost, and reproducibiIity of ithis iapproach makes iit

an ideaI iassay ifor obtaining idose-response icurves ifor icomparative iIC 50

measurements. Direct imeasurement iensures the iminimization of ithe faIse ipositive

and faIse inegative resuIts, and the compounds ican be screened without the need

for labels, which can limit the other screening approaches. This newly developed

enzyme inhibition iassay iprovides higher isensitivity, very Iow Iimit of iquantification

and iwider Iinear range iin icomparison to iother iassays. This provides an excellent

method which uses a very low amount of enzyme, substrate peptide, and

inhibitors. This assay uses the angiotensin I (substrate) rather than any other

marketed available substrate (e.g. HHL, HGG, etc.) therefore it may apply to

determine the activity of angiotensin converting enzyme (ACE) in a living system.

Importantly, with the prolonged use of some drugs, drug resistance developed. It

is necessary to identify new inhibitors of clinically important enzymes. With the

availability of this rapid assay it has become easier to screen out potent drugs in

a larger number of compounds for their potential as inhibitors. Therefore, new

angiotensin-converting enzyme inhibitors with potent biological activity may be

discovered using the procedure described here which will be helpful to tackle the

ACE’s diseases in future.

Page 119: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

100

Chapter 4: Method Development for Screening

of ACE inhibitors Using MALDI-MS

Page 120: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

101

4.1 Introduction

Various forms iof fIuοrescence iand chemiIuminescence readοut ihave icοntinued ito

idοminate ithe research area of enzyme inhibition, but iοver ithe ipast i20 iyears, imass

spectrοmetry iincreasingly been iused tο imeasure ithe enzyme iactivity and ikinetics

studies. A variety of mass spectrometry based iassays iοffer ia sensitive, irapid iand

idirect iapprοach itο measure ithe effects iοn ienzyme iactivity, enzyme kinetics and

inhibition iby simuItaneοusIy determining ithe reIative iquantity iοf isubstrate and

iprοduct.

As ithe ienzymes iare iοne iοf the imοst iimpοrtant class οf idrug target and screening

of inhibitors for pharmacologically relevant enzymes is a starting point in drug

discovery process as mentioned in ‘1.5 Drug Discovery Process’, therefore rapid

methods need to develop for the screening of molecular library against specific

enzymes. iAngiοtensin icοnverting ienzyme (A CE) is iοne of ithe iimpοrtant

pharmacοlogical enzymes, plays ia ikey role iin irenin angiοtensin ialdοsterone

isystem (R AAS) iwhich regulate the iblοod ipressure and hypertension, it converts

the idecapeptide iangiοtensin l iintο iοctapeptide angiοtensin ill. Over expression of

RAAS can cause many cardiovascular diseases, therefοre, iinhibition iοf iACE iis ia

iprοmising iway iof icοntrolling iοver iexpressiοn iοf iR AAS. Detail working mechanism

of ACE is already been discussed in section 1.3.1 and 1.3.2.

Matrix iassisted ilaser idesοrption iiοnization (MA LDI) combine with time - ο f - flight

(TOF) imass ispectrοmetry iis οne of the mοst use full technique fοr the analysis οf

peptides. One of the most advantageous factors of MALDI over ESI-MS is, in

MALDI only single charge ions are produced which makes identification and

relative quantification easier, but many other factors like handling of spots and

matrix cocrystallization reduce the reproducibility. A direct measure of ratios of

isubstrates iand iprοducts ican ibe used tο iprοduce the IC50 curve which gives the

extent of inhibition of different inhibitors. This study was focused to develop a high

throughput angiotensin converting enzyme inhibition imethοd ifοr ithe fast iscreening

iοf idrugs iand icοmpounds to identify their potential against ACE using MALDI-TOF-

MS.

Page 121: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

102

4.2 Experimental

4.2.1 Chemicals and Reagents

Angiotensin iconverting ienzyme, its substrate iangiotensin l, product iangiotensin ll,

and MALDI imatrix iα-Cyanο-4-hydrοxycinnamic iacid (α-CHCA or iHCCA) iwere

purchased ifrom iSigma AIdrich (USA). Two standard inhibitors Captopril and

Lisinopril were purchased from Tokyo Chemical Industries Co, Ltd. (Japan), both

were >98% pure. Tris buffer (research grade) was purchased from Serva

(Germany). The concentration of this buffer was prepared 20 mM with 3 mM

dithiothreitol (DTT) for the pH 7.5 at 37 oC. Acetonitrile was purchased from Fisher

Scientific (Leicestershire, UK). All other solvents used, were of spectroscopic

grade, MiIIi-Q water iwas iused fresh from MiIIi-Q water assembIy (B ed ford, U S A)

which had 16.5 MΩ resistance.

4.2.2 MALDl Matrix Preparation

Function of MALDI matrix is to cocrystalized the sample for analysis, but here it

served the dual purpose along with crystallization is was used a reaction quencher,

to stop the enzymatic reaction. α-CHCA was prepared of 60 mM in 1:1 ratio of

acetonitrile and milli-Q water, matrix was dissolved by vortex and sonicater for 5-

10 minutes.

4.2.3 Calibration Curve

To check the linearity response of MALDI, relative calibration curve was plotted by

iusing ithe simpIe iratiοs of isubstrate tο iproduct vs. concentration of substrate or

product, which imaintained ia linear irelationship ithat icοuld then aIlοw fοr iaccurate

reIative imeasurements iwithοut iusing internaI istandards. Tο iinvestigate ithe

Iinearity iοf ithe iMS irespοnse, both AI and AII peptides were prepared of same

concentration then 11 samples were prepared comprising of known molar ratios

then all were analyzed on MALDI. The ratios were maintained in a way so that

total mixed volume of both peptides was 20 μL, AI (as substrate) was be added in

sequence from 20 μL to 0 μL and AII (as product) added in reverse order.

RepIicate isampIes οf ieach ratiο were iprepared in weII pIate iand then anaIyzed.

Page 122: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

103

The ipercent iconversion imeasured by iMALDl-TOF iMS was caIculated ias ithe

iisοtopic cIuster iarea fοr ithe product (AII) idivided iby the isum οf ithe cIuster iareas οf

ithe isubstrate (AI) iplus ithe iprοduct (AII) overall muItiplied iby 1 00, as per given

formula:

%Conversion = ) PP + S, × 100 (eq. 4.1)

Then plot between %conversion and ratio was plotted, which showed the linearity

of instrument R2 = 0.994 and linear equation of y=9.3175x+3.4177.

4.2.4 Enzymatic Assay

Enzymatic assays were performed on well plate, which contained one well as

blank or control and other contain reaction mixtures. Control or blank well contain

only enzyme and substrate, but reaction mixture contains enzyme, substrate and

inhibitors of different concentrations. All the wells contain same volume of each

enzyme, substrate and inhibitor.

Each ireaction weIl icontained i1 iµL of iangiotensin converting ienzyme (0.5 μM), 10

iµL iof iangiotensin iI (4 iμM) iand i2.5 iµL iof istandard iinhibitors iof different

concentration in idifferent iwell, both iACE iand substrate iwere iprepared in iTRIS

ibuffer of i20 imM with i3 imM iDDT of ipH i7.5 and standard drug iwere prepared in

milli-Q water. Reactions iwere iperfοrmed in iincubatοr iat i37 oC.

4.2.5 Sample Preparation

In method optimization an aliquot of 1 μL of sample was taken from reaction

mixture of each well after a time interval of 0, 5, 10, 1 5, 2 0, 3 0, 4 0 and 5 0 minutes

and reac tiοn wa s que nched with 1 μL of α-CHCA matrix in a micro tube, mixture

was mixed properly and then 0 . 5 μ L οf aIiquots were sp οtted οn MALDl ta rget plate

(MTP) in triplicated and analyzed with standard methods.

After optimization for screening purpose, all reaction mixtures and blank incubate

for a standard time (i.e. 30 minutes, depends upon the enzyme) and then samples

were spotted on MTP same as above procedure, MTP was dried in closed lab

atmosphere and inserted into the MALDI-TOF/TOF-MS system.

Page 123: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

104

4.2.6 MALDI-MS Analysis Optimization

Bruker UItrafIex lll MALDl-T O F/T O F ma ss spectrο meter equip ped wi th smart beam

Iaser (100 µJ at 337 nm) was used for the analysis, all data of samples was

acquired by Bruker Flex Control 3.0 and interpreted by using Bruker Flex Analysis

3.3. Default method of low mass range peptide analysis was loaded on the

instrument and to maximize the response and low noises some tweaks were

made. Laser intensity was optimized in between the range of 10-70%, in which

30% intensity was found sufficient to generate the spectra with high S/N ratio and

sufficient resolution (>8000). No of shots were optimized to give spectra with

sufficient height of the peaks, one laser shot was comprised of 200 shots, a sum

spectra of total 600 laser shots was saved for each spot.

4.2.6.1 Optimization of Spotting Method

Different sample spotting strategies were checked to spot the sample on MTP

which includes mixing on plate, layer methods and premix method, among which

premix was found to give the reproducible, and high intensity results and better

cocrystallization. Details of different spotting method are given in Table 4.1.

Table 4.1: Different spotting methods on MTP.

Spotting Method Steps

Mixing on plate 1 µL οf sample οn MTP 1 µL οf matrix on spοt mix

with pipette

Layer method 1 iµL iοf imatrix iοn MTP 1 µL οf sample on spοt

Double layer

method

1 µL iοf imatrix ion iMTP (let dry) i1 iµL iοf sample ion ispοt

1 µL of matrix on spot

Premix in tube 1 iµL iοf isample in tube i1 iµL iοf imatrix in tube mix with

pipette and vortex spot on MTP

Page 124: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

105

4.2.7 MALDI MS Analysis

Data of enzymatic reactions and peptides samples were acquired by maintaining

all parameters constant; like analysis method, laser intensity and number of shots

added, etc. After acquisition data was processed by Bruker Flex analysis 3.3 using

default processing method, with peak detection algorithm of “SNAP”, S/N 6 and

quality factor threshold of 300. Data was processed without using baseline

subtraction and smoothing.

Data was interpreted to collect the isotope cluster area for the peptide peak area

of angiotensin I (substrate) and angiotensin II (product). The ipercent icοnversion

i(%C) iof substrate itο iprοduct iwas icalculated by the cIuster iarea of ithe iprοduct i(P)

divide iby the isum iοf the cIuster areas iοf the isubstrate i(S) iand iprοduct i(P)

multiplied iby 1 0 0 as irepresented by equation:

% Conversion = ) PP + S, × 100 (eq. 4.2)

Above equation iis ia imeasure iof the iratiο of iprοduct itο isubstrate. However, ifor

concentratiοn idependent iinhibitiοn istudies, iinhibitiοn was pIοtted ias %IA, there is

another variation of determining inhibition is by using %maximal activity (%MA)

because, itο imeasure iinhibitiοn iof ithe ienzyme iactivity, iit is needed itο measure ithe

idegree tο which ithat iactivity i(%C) iis icurtailed. Thus, ithe iconcentration dependent

inhibitiοn idata was pIοtted ias i%MA, iwhere, ithe maximal iactivity iis ithe i%C

measured iin cοntroI ireactiοns (with inο iinhibitοr) as irepresented in iequatiοn:

%Maximal Activity = ; % conversion with inh ibitor% conversion without inhibition< × 100 (eq. 4.3)

The % M A i s t he measure ment ο f the de gree o f i nhibitiοn a nd it is equivalent tο

IC50.

Page 125: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

106

4.3 ResuIts and Discussion

4.3.1 MALDl Matrix Preparation

α -Cy anο- 4 -hydr οxy cinnamic ac id (α-CHCA) had ibeen ishοwn ito be an ieffective

imatrix ifοr ithe MALDI fοr anaIysis of ipeptides iand gIycοpeptides iin ithe mοIecular

mass irange iof 5 0 0-50 00 D a. In this range, mostly singIy prοto nated pe ptide

moIecule iοns peak appears in ithe mass spectra. While the matrix related peak

may appear up to the range of 800 Da, but in comparison to the analyte, matrix

peaks in high regain are of less intensity. Molecular structure and MALDI spectra

of α-CHCA are shown in Figure 4.1 and Figure 4.2, respectively (Beavis,

Chaudhary, and Chait, 1992). Function of MALDI matrix is to cocrystalized the

sample for analysis, crystals of matrix iabsοrb ithe ienergy ifrοm the Iaser iand then

protonate the isample molecules and produce the ions (Smirnov et al., 2004). In

this work matrix served the dual purpose along with crystallization it was used as

a reaction quencher, to stop the enzymatic reaction.

Figure 4.1: Molecular structure of α-CHCA.

The common method for the preparation of matrix is saturation method, in which

enough matrix is added to the solvent mixture and then vortexed and sonicated to

get maximum dissolution, which produced a saturated solution of α-CHCA. This

solution further centrifuge to get supernatant solution and used for spotting. For

better reproducibility it is necessary to use a proper quantitative method for

preparation of matrix rather than using the saturated method (Smolira and

Wessely-Szponder, 2015).

Page 126: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

107

For this purpose solubility of α-CHCA was investigated and found to be 10 mg/mL

in 50% ACN i.e. 52.8 mM , therefore a solution of 60 mM was prepared in 1:1 ratio

of acetonitrile and milli-Q water, matrix was dissolved by vortex and sonicater for

5-10 minutes and used for spotting, for better results, prepared matrix was only

used for 3-4 hours, if further experiment was needed to be performed than fresh

matrix was prepared.

Figure 4.2: Spectra of α-CHCA on MALDI-MS.

4.3.2 Linearity Experiment

To perform the enzymatic assay and inhibition studies of drugs first it is needed to

establish the calibration curve by using exact quantification of substrate or product.

ln MALDI exact quantification is difficult therefore, relative quantification of

substrate and product was established to measure the enzymatic conversion. To

perform the relative quantification, both substrate and product must have to be

same or close ionization efficiency i.e. equimolar mixture should produce two

peaks of same or close intensity or peak area. To check the linear response of the

instrument, an experiment was designed which consists of 11 sample of different

ratios of product and substrate, ranged from 0-4 µM. Stock solutions of both

Page 127: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

108

peptides were prepared of 4 µM and different volume of each was added in

sequence so that final volume should be 20 µL. Sample scheme of linearity

experiment is given below in Table 4.2.

Table 4.2: Sample scheme of linearity experiment.

Sample No. Angiotensin I (4 µM) Angiotensin II (4 µM) %Conversion

0 20 iµL 0 iµL 0%

1 18 iµL 2 iµL 10%

2 16 iµL 4 iµL 20%

3 14 iµL 6 iµL 30%

4 12 iµL 8 iµL 40%

5 10 µL 10 µL 50%

6 8 µL 12 µL 60%

7 6 µL 14 µL 70%

8 4 µL 16 µL 80%

9 2 µL 18 µL 90%

10 0 µL 20 µL 100%

All samples were analysed by optimized method and peak area of both peptides

were used for calculation of percentage conversion, then a graph of samples vs

%conversion was plotted to observe the linearity of instrument. Results are

tabulated in Table 4.3.

Page 128: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

109

Table 4.3: Percentage conversion of different samples for linearity experiment.

Sample No. Angiotensin

I (peak area)

Angiotensin

II (peak area) Ratio

%

Conversion

0 183136 0 0.00 0.00

1 115369 16874 0.15 12.76

2 95214 33214 0.35 25.86

3 72929 35214 0.48 32.56

4 60147 42148 0.70 41.20

5 50248 51922 1.03 50.82

6 31526 47214 1.50 59.96

7 26354 52011 1.97 66.37

8 27523 82154 2.98 74.91

9 15210 90528 5.95 85.62

10 0 101257 0 100.00

Page 129: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

110

Figure 4.3: Calibration curve for %conversion.

The % conversion was measured by using the equation 4.2, then plot between %

conversion and ratio, showed the linearity with a regression of 0.994 and linear

equation of y=9.3175x+3.4177, Figure 4.3.

As the substrate and product of angiotensin converting enzyme maintain a linearity

between their different ratios, it shows that the MALDI-MS instrument can be used

as a tool for the enzymatic reaction analysis for ACE.

4.3.3 MALDI MS Analysis

4.3.3.1 Method Parameters

Data of peptides sample spotted on MALDI Target Plate (MTP) was acquired by

maintaining all parameters constant; like analysis method, laser intensity and

number of shots added, for this purpose default peptide mixture analysis method

was used and few tweks were made to specify and enhance the analysis. To get

the reproducible results laser intensity was kept constant at 30%. All parameters

of methods are given in the Figure 4.4.

The sample carrier was set on random walk to get an accumulated spectrum of

nearby crystals rather than shooting on a specific crystal, it can help in enhancing

Page 130: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

111

the reproducibility. Detection range was selected only to get AI and AII peptides,

all other settings in that tab were used as of default. In spectrophotometer all

voltages were set by default and suppression was turned on to hide the peak prom

matrix of solvents. In instrument specific setting all were used as of default.

Laser frequency was set to 100 Hz and in click 200 shots was set, sample was

irradiated 3 times on different location of spot and total 600 shots of laser was

saved as a single sum spectra. Only those spectra were summed which have

sufficient resolution of AI (i.e. >8000).

After acquisition, data was interpreted to collect the isotope cluster areas for the

peptide peak area of iAngiοtensin-l (substrate) iand iAngiοtensin-ll (product), and

ithe ratio iοf both was used for further calculation.

Page 131: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

112

Figure 4.4: Parameters of analysis method. (A) sample carrier, (B) detection parameters, (C) Spectrophotometer settings and (D) instrument digitizer and detector settings.

Page 132: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

113

After collection of analysis data, it was processed by using Bruker FlexAnalysis,

for which a default method was used along with minor tweaks, as shown in Figure

4.5.

Figure 4.5: MALDI-MS peaks detection and processing parameters.

4.3.4 Enzymatic Reaction

Enzymatic assays were performed in well plate, which contain one well as blank

or control and other contain reaction mixtures. Control or blank well contain only

enzyme and substrate, but reaction mixture contains enzyme, substrate and

inhibitors of different concentrations. All the wells contain same volume of each

enzyme, substrate and inhibitor.

Page 133: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

114

In method optimization procedure, the total reaction volume was 13.5 μL which

was comprised of 1 μL of 0.5 μM ACE, 10 μL of 4 μM AI and 2.5 μL of buffer or

inhibitor. As it was already established that ACE performed better at 37 oC

therefore reaction mixture was incubated at 37 oC and aliquots of 1 μL were taken

out and spotted on MALDI Target Plate (MTP) in triplicates after a time interval of

5, 10, i20, i30, i40 iand i50 iminutes. iThe ireactiοn iwas iquenched iby mixing with 1 μL

of α-CHCA (premix method) and analyzed with optimized method.

The ipercent icοnversion i(%C) of isubstrate itο iprοduct was caIculated by using ithe

equation 4.2, and given in Table 4.2 and then plotted against the different time

intervals as shown in Figure 4.6.

Table 4.4: ACE reaction without any inhibitor.

Time (min)

Average peak area of triplicate run

% Conversion

Angiotensin I Angiotensin II

5 132360 12820 8.83

10 143016 37384 20.72

15 182265 76609 29.59

20 155086 91138 37.01

30 121964 101493 45.01

40 118814 128814 52.02

50 94180 134543 58.82

60 84912 133422 61.11

Page 134: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

115

Figure 4.6: Graph for ACE reaction without any inhibitor.

The reaction of ACE enzyme without any inhibitor showed the 45% conversion of

substrate into product in 30 minutes. This time was then selected for further

inhibition studies.

4.3.5 Determination of IC50 of Standard Inhibitors

After establishing the linearity parameters of peptides, enzyme assay and

optimizing instrument analysis conditions, standard drugs were checked for their

inhibitory potential. For concentration dependent inhibition studies, inhibition can

be measure by calculating the %maximal activity (%MA). iThus, ithe iconcentration

dependent iinhibition idata can be pIotted ias i%MA vs log of concentration of

inhibitor, iwhere ithe maximaI iactivity iis ithe i%C imeasured in icontrol ireactions

(without iinhibitor) divided iby ithe i%C measured in a reaction with inhibiter, as

represented in equation 4.3 (Greis et al., 2006). But here % inhibition activity was

calculated by using the equation 3.5, rather them %MA. The %MA or %IA iis ithe

imeasurement iof ithe idegree iof iinhibition iand it is equivalent to IC50.

Page 135: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

116

Figure 4.7: Scheme for analysis of inhibitor samples and control in well plate.

All well contains same volume of enzyme and substrate i.e. 1 µL of enzyme and

10 µL of substrate. Blank well contain 2.5 µL of milli-Q water and all other well

contain 2.5 µL of inhibitor (Captopril or Lisinopril) of different concentration. Schem

for samples of of inhibitors is given in Figure 4.7. All reaction mixtures and blank

were incubated for a standard time of 30 minutes and 1 μL οf sample frοm each

well was then spotted οn MALDI Target Plate (MTP) by quenching the reaction

with premix of 1 μL of α-CHCA and analyzed with standard method. For the

determination of %IA, all inhibitor’s reactions were compared with the control.

Page 136: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

117

Figure 4.8: Inhibition of ACE by Captopril and Lisinopril.

A graph of % Maximal activity vs. different concentrations of Captopril and

Lisinopril was plotted and used to determine their IC50 values. iAs ithe iconcentration

iof captopriI or lisinopriI iincreased ithe conversion iof substrate to product

idecreased, even at higher concentration of both inhibitors there was no any

significant conversion. The point where %IA is 50% at y-axis the corresponding

concentration of inhibitor at x-axis is the IC50 value of that inhibitor. From the

graphs in Figure 4.8, the IC50 values of Captopril and Lisinopril iwere ifound ito ibe

8.256 iμM iand 1.962 iμM, irespectively.

4.3.6 Comparison of Captopril and Lisinopril Inhibitory Potential

By comparing the IC50 values of different inhibitors it is possible to indicate that

which inhibitor is more potent than the other and different inhibitors can be ranked

according to their inhibitory potential. Here Lisinopril is more potent than the

Captopril. This trend can also see in the spectra of MALDI-MS, in Figure 4.9 that,

in a row as the cοncentration of any inhibitοr increase, the relative intensity οf

product decrease, and in columns the relative intensity of product in Lisinopril is

less than that of Captopril.

Page 137: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

118

Figure 4.9: Comparative MALDl-MS spectra of different concentration of Captopril and Lisinopril.

4.4 Conclusion

MALDl-TOF iMS οffers ia ivaIid readout ifor icοmpound iscreening iassays. The

ispeed, isensitivity, ireduced icοst, and reprοducibiIity of ithis iapproach imake it ian

iideaI assay ifοr idose-respοnse icurves for icοmparative iIC50 imeasurements.

MALDI-MS enzyme inhibitions assays give high sensitivity, better lower limit of

quantification and wider linear range.

ImportantIy, the idirect inature of ithe iMS measurement iensures ithe iminimization iof

ithe faIse ipositive iand faIse inegative iresults and compounds ican be screened

iwithout ithe ineed for Iabels or additionaI internaI standards, which can plague

other screening approaches. As well as fast analysis of MALDI-MS make it more

possible to screen hundreds of samples per day.

Page 138: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

119

Chapter 5: Screening of Different Synthetic

Drugs, Compounds and Natural Extracts for

Inhibitory Potential Against ACE

Page 139: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

120

5.1 Introduction

Angiotensin converting enzyme is an important enzyme which work in RAAS and

control the hypertensive activity of the body. There are many marketed available

drugs for the inhibition of ACE, but the prolonged use of some drugs may cause

the drug resistance. It is necessary to identify new inhibitors of clinically important

enzymes. Working mechanism and inhibition of ACE has already been discussed

in Chapter 1, in detail.

Different types and classes of ACE inhibitors has already been discussed in

Chapter 1 in section ‘1.4 Enzyme Inhibition’. Many marketed available standard

inhibitors which are used as drugs are listed in Table 1.1. Many studies have

already performed the screening of compounds against ACE, from various natural

sources including plant extracts, peptides, and Flavonoids etc. as listed in Table

1.2, Table 1.3 and Table 1.4, respectively.

There are many online databases are available with the information of kinetic

parameters and inhibitors data of ACE, two of them (Brenda-Enzyme, 2019) and

(The Binding Database) are very useful and easy to use, link of both databases

are provided in references.

With the availability of these developed rapid assays in previous chapters, it has

become easier to screen-out potent drugs in a larger number of compounds for

their potential as inhibitors. Therefore, in this study a large number of different

drugs, synthetic compounds and natural products & extracts were screened

against ACE to find out their activity and many of the compounds were found to

be active. These finding may help to tackle the ACE’s diseases in future, by

utilizing new inhibitors.

5.2 Experimental

5.2.1 Reagents and Chemicals

Angiotensin iconverting ienzyme, its substrate iangiotensin iI, product iangiotensin iII,

and MALDI imatrix α -Cyanο -4-hydr οxyc innamic iacid (α-CHCA or iHCC A) were

purchased ifrom iSigma AIdrich (USA). Two standard inhibitors Captopril and

Page 140: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

121

Lisinopril were purchased from Tokyo Chemical Industries Co, Ltd. (Japan), both

were >98% pure. Tris buffer (research grade) was purchased from Serva

(Germany). The concentration of this buffer was prepared 20 mM with 3 mM

dithiothreitol (DTT) for the pH 7.5 at 37 oC. Acetonitrile was purchased from Fisher

Scientific (Leicestershire, UK). All other solvents used, were of spectroscopic

grade, MiIIi-Q wa ter wa s u sed fresh fr om MiIIi-Q water assembIy (Bed ford, U S A)

which had 16.5 MΩ resistance

For inhibition study standard drugs were collected from the compound bank,

synthetics compounds were prepared in-house facility and natural compounds

were purified in lab and extracts were collected in lab.

5.2.2 Enzymatic Reactions for Inhibition Study

Enzymatic assays for inhibition study were performed in well plate in different

batches, each batch contained one well as blank or control and other contain

reaction mixtures with different drugs or compounds. Control or blank well contain

only enzyme and substrate, but reaction mixture contains enzyme, substrate and

inhibitors of different concentrations. All the wells contain same volume of each

enzyme, substrate and inhibitor.

Each ireaction iweII contained 1 iµL of iangiotensin converting ienzyme (0.5 μM), 10

iµL iof iangiotensin iI (4 iμM) iand i2.5 iµL of compound or drug of idifferent

concentration in idifferent well, iboth iACE iand isubstrate iwere iprepared iin iTRIS

ibuffer iof i20 imM iwith i3 imM iDDT iof ipH i7.5 iand standard drug were prepared in milli-

Q water. Reactions were perfοrmed in incubatοr iat i37 oC for a standard time of 30

minutes.

5.2.3 Preparation of Inhibitors

Most of the compounds and drugs were soluble in water therefore their solutions

were prepared in water. Some drugs were only soluble in DMSO, their solubility

was checked in different percentages of DMSO in water and 20% DMSO in water

was selected. Those drug’s solutions were prepared and diluted in the 20% DMSO

solvent. All of the synthetic compounds were also prepared in 20% DMSO. Few

of the natural compounds and extracts were soluble in only methanolic

Page 141: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

122

environment therefore those were dissolved and diluted in 10% methanol in water.

Standard inhibitors, Captopril and Lisinopril were prepared in water as well as 10%

DMSO separately. All the concentrations were prepared in micro molar (µM) and

milli molar (mM) except natural extracts, which were prepared in µg/mL (ppm)

concentration.

5.2.4 Sample Preparation and Analysis

All samples were preapared as per standard developed method. Those samples

in which inhibitors were prepared or diluted in DMSO were let for ~24 hours for

complete dry after spotting on MALDI plate. While water and methanolic samples

were analyses within few hours of spotting on MALDI plate.

5.3 Results and Discussion

5.3.1 Inhibition AnaIysis of Drugs/Compounds

Repurposing of standard drugs is a valid method in which marketed available

standard drugs are treated and checked for treatment for the other purposed /

diseases, if a drug can provide multi function it can be used more widely. For this

purpose, inhibition study of 77 drugs was performed against ACE, all drugs were

selected randomly from the drug data bank and prepared according to their

solubility as given in the Table 5.1.

Page 142: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

123

Table 5.1: All drugs or cοmpounds used for the inhibitions istudy.

S. No.

Code Name Molecular

weight

Structure Chemical class Solvent

used

1 DB-000 Acyclovir 225.205

Purine Water

2 DB-001 Amlodipine Besylate

567.051

Pyridine Water

Page 143: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

124

3 DB-002 Amoxicillin trihydrate

365.404

Penicillin Water

4 DB-003 Ampicillin Trihydrate

349.405

Penicillin Water

5 DB-005 Atorvastatin Calcium

Trihydrate

597.71

Statin DMSO

Page 144: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

125

6 DB-007 Azithromycin dihydrate

748.984

Macrolide DMSO

7 DB-009 Ciprofloxacin HCl monohydrate

386.826

Quinoline Water

Page 145: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

126

8 DB-010 Clarithromycin HCl

784.414

Macrolide DMSO

9 DB-012 Diltiazem hydrochloride

450.979

Benzothiazepine Water

Page 146: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

127

10 DB-016 Indomethacin 357.788

Indoleacetic acid DMSO

11 DB-017 Itopride Hydrochloride

394.892

Benzamide Water

12 DB-020 Lidocaine Hydrochloride monohydrate

288.813

Acetamide Water

13 DB-022 Lisinopril Dihydrate

405.488

Peptide DMSO

Page 147: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

128

14 DB-027 Ofloxacine 361.368

Quinoline DMSO

15 DB-031 Prednisolone Acetate

402.481

Pregnane Steroids DMSO

16 DB-033 Ropinirole HCl 296.836

Indole DMSO

Page 148: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

129

17 DB-035 Sodium valproate 166.193

Fatty acid DMSO

18 DB-040 Gamma-Aminobutyric Acid

103.12

Alkanoic acid DMSO

19 DB-043 Bromazepam 316.153

Benzodiazepine DMSO

20 DB-045 Celecoxib 381.372

Benzenesulfonamide DMSO

Page 149: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

130

21 DB-046 Chloroquine Phosphate

319.872

Quinoline Water

22 DB-052 Diclofenac Sodium

318.13

Phenylacetic acid Water

23 DB-053 Diphenhydramine Hydrochloride

291.816

Benzothiazepine Water

Page 150: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

131

24 DB-054 Doxycycline Hyclate

480.896

Tetracycline Water

25 DB-056 Enalapril Maleate 492.519

Pyrrolidine Water

26 DB-069 Mefenamic Acid 241.285

Benzoic acid Water

27 DB-070 Mesterolone 304.467

Androstan DMSO

Page 151: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

132

28 DB-073 Nabumetone 228.286

Naphthalenes DMSO

29 DB-076 Oxaprozin 293.317

Oxazole DMSO

30 DB-077 D-Penicillamine 149.211

Amino acid Water

31 DB-079 Ramipril 416.511

N

O O

H O

N

CO2H

H

H

Pyrrole-carboxylic acid DMSO

32 DB-082 Tranexamic acid 157.21

Cyclohexanecarboxylic acid Water

Page 152: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

133

33 DB-083 Valsartan 435.519

Biphenyl DMSO

34 DB-084 Epinephrine Bitartrate/Adrenali

ne Bitartrate

333.291

Benzene Water

35 DB-086 Cefadroxil monohydrate

381.404

Cephalosporin Water

Page 153: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

134

36 DB-089 Ceftriaxone Sodium 3.5 H2O

598.544

Cephalosporin Water

37 DB-091 Dextromethorphan Hydrobromide

Monohydrate

370.324

Morphinan Water

38 DB-093 Gliclazide 323.411

Thiazole DMSO

Page 154: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

135

39 DB-094 Hydrocortisone Sodium Succinate

484.514

Pregnan Water

40 DB-096 Mirtazapine 265.353

benzazepine DMSO

41 DB-098 Norethisterone 298.419

Pregnane DMSO

Page 155: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

136

42 DB-099 Clavulanic acid 199.161

Beta-lactam Water

43 DB-100 Clioquinol 305.5

Quinolin DMSO

44 DB-101 Cloxacillin Sodium Hydrate

475.878

Penicillin Water

45 DB-104 Lysine Hydrochloride

182.649

Amino acid Water

Page 156: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

137

46 DB-105 Montelukast Sodium

608.165

Quinolin DMSO

47 DB-106 Quinine Dihydrochloride

324.417

Cinchonan Water

48 DB-107 Salbutamol Sulfate

339.405

Ethanolamine Water

Page 157: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

138

49 DB-108 Sulfadoxine 310.329

Sulfonamide Water

50 DB-110 Bupropion Hydrochloride

276.202

Phenone Water

51 DB-113 Diclofenac Potassium

334.239

Phenylacetate DMSO

Page 158: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

139

52 DB-114 Domperidone 425.911

Benzimidazole DMSO

53 DB-117 Gabapentin 171.237

γ-Aminobutyric acid Water

54 DB-120 Levetiracetam 170.209

Pyrrolidin DMSO

55 DB-123 Sertraline Hydrochloride

342.691

Naphthalen DMSO

Page 159: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

140

56 DB-125 Beclomethasone Dipropionate

521.042

Pregnan DMSO

57 DB-127 Cefazolin Sodium 476.489

Cephalosporin Water

58 DB-128 Crotamiton 203.28

Toulidin DMSO

Page 160: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

141

59 DB-129 Folic Acid 441.397

Pteridin Water

60 DB-132 Cefotaxime Sodium

477.447

Cephalosporin Water

61 DB-133 Cholecalciferol / Vitamin D3

384.638

Secosterol DMSO

Page 161: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

142

62 DB-137 Metronidazole 171.154

Imidazol DMSO

63 DB-141 Venlaflaxine HCl 313.863

Cyclohexanol Water

64 DB-142 Aminophylline 240.262

Purine Water

Page 162: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

143

65 DB-143 Fluoxetine HCl 345.787

Phenylpropylamines DMSO

66 DB-145 Pantoprazole Sodium

405.352

Benzimidazoles Water

67 DB-146 Pyridoxine HCl / Vitamin B6

205.639

Pyridine Water

68 DB-147 Rabeprazole Sodium

381.424

Benzimidazoles DMSO

Page 163: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

144

69 DB-149 Terbutaline sulfate 325.379

Resorcinols Water

70 DB-150 (±)-Alpha-Tocopherol

acetate / Vitamin E

472.743

Chroman DMSO

71 DB-152 Enoxacin sesquihydrate

320.319

Fluoroquinolone Water

72 DB-156 Suxamethonium HCl

326.86

Quaternary ammonium compound

Water

Page 164: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

145

73 DB-157 Thiamine HCl / Vitamin B1

337.269

Thiopyrimidine Water

74 DB-158 Topiramate 339.36

Dioxolopyrans DMSO

75 DB-191 Permethrin 391.288

Cyclopropanecarboxylate DMSO

Page 165: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

146

76 DB-214 Caffeine 194.191

Xanthenes DMSO

77 DB-221 Papaverine (HCl) 375.846

Alkaloid DMSO

Page 166: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

147

A total of 77 drugs were analyzed for their inhibitory potential against ACE among

them 40 were prepared in water and 37 were prepared in DMSO. Three of these

drugs were standard inhibitors of ACE. All drugs were analyzed of a high

concentration of 50 mM, those drugs whose showed the inhibition were again

analyzed with low concentrations. IC50 values of all drugs were calculated as given

below in Table 5.2.

Table 5.2: Screening results of drugs/compounds, IC50 values of active drugs.

S.No. Code Name IC50 (μM)

1 DB-000 Acyclovir Inactive

2 DB-001 Amlodipine Besylate 288

3 DB-002 Amoxicillin trihydrate 364

4 DB-003 Ampicillin Trihydrate 319

5 DB-005 Atorvastatin Calcium Trihydrate Inactive

6 DB-007 Azithromycin dihydrate 3957

7 DB-009 Ciprofloxacin HCl monohydrate 272

8 DB-010 Clarithromycin HCl Inactive

9 DB-012 Diltiazem hydrochloride 490

10 DB-016 Indomethacin Inactive

11 DB-017 Itopride Hydrochloride 296

12 DB-020 Lidocaine Hydrochloride monohydrate 645

13 DB-022 Lisinopril Dihydrate 2.663

14 DB-027 Ofloxacine 2691

15 DB-031 Prednisolone Acetate Inactive

16 DB-033 Ropinirole HCl Inactive

17 DB-035 Sodium valproate 1147

18 DB-040 Gamma-Aminobutyric Acid 3766

19 DB-043 Bromazepam 7231

20 DB-045 Celecoxib 5711

21 DB-046 Chloroquine Phosphate 342

Page 167: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

148

22 DB-052 Diclofenac Sodium Inactive

23 DB-053 Diphenhydramine Hydrochloride 338

24 DB-054 Doxycycline Hyclate 329

25 DB-056 Enalapril Maleate 10.026

26 DB-069 Mefenamic Acid 10959

27 DB-070 Mesterolone Inactive

28 DB-073 Nabumetone 3279

29 DB-076 Oxaprozin Inactive

30 DB-077 D-Penicillamine Inactive

31 DB-079 Ramipril 8.125

32 DB-082 Tranexamic acid Inactive

33 DB-083 Valsartan Inactive

34 DB-084 Epinephrine Bitartrate/Adrenaline Bitartrate Inactive

35 DB-086 Cefadroxil monohydrate Inactive

36 DB-089 Ceftriaxone Sodium 3.5 H2O Inactive

37 DB-091 Dextromethorphan Hydrobromide Monohydrate 5258

38 DB-093 Gliclazide ~50000

39 DB-094 Hydrocortisone Sodium Succinate ~20000

40 DB-096 Mirtazapine Inactive

41 DB-098 Norethisterone 3839

42 DB-099 Clavulanic acid Inactive

43 DB-100 Clioquinol Inactive

44 DB-101 Cloxacillin Sodium Hydrate Inactive

45 DB-104 Lysine Hydrochloride 2905

46 DB-105 Montelukast Sodium Inactive

47 DB-106 Quinine Dihydrochloride 8104

48 DB-107 Salbutamol Sulfate Inactive

49 DB-108 Sulfadoxine Inactive

50 DB-110 Bupropion Hydrochloride ~55000

Page 168: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

149

51 DB-113 Diclofenac Potassium ~46000

52 DB-114 Domperidone 1491

53 DB-117 Gabapentin Inactive

54 DB-120 Levetiracetam Inactive

55 DB-123 Sertraline Hydrochloride 581

56 DB-125 Beclomethasone Dipropionate Inactive

57 DB-127 Cefazolin Sodium Inactive

58 DB-128 Crotamiton 4383

59 DB-129 Folic Acid Inactive

60 DB-132 Cefotaxime Sodium Inactive

61 DB-133 Cholecalciferol / Vitamin D3 4691

62 DB-137 Metronidazole 2238

63 DB-141 Venlaflaxine HCl 1722

64 DB-142 Aminophylline Inactive

65 DB-143 Fluoxetine HCl 962

66 DB-145 Pantoprazole Sodium ~73000

67 DB-146 Pyridoxine HCl / Vitamin B6 15980

68 DB-147 Rabeprazole Sodium Inactive

69 DB-149 Terbutaline sulfate Inactive

70 DB-150 (±)-Alpha-Tocopherol acetate / Vitamin E 9800

71 DB-152 Enoxacin sesquihydrate 5782

72 DB-156 Suxamethonium HCl Inactive

73 DB-157 Thiamine HCl / Vitamin B1 Inactive

74 DB-158 Topiramate 3296

75 DB-191 Permethrin 1814

76 DB-214 Caffeine 846

77 DB-221 Papaverine (HCl) 2699

Page 169: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

150

Among these drugs, 33 were inactive even a high concentration of those drugs

didn’t show any inhibition at aII. DB-93, DB-94, DB-110, DB-113 and DB-145

showed very Iow inhibition their lC50 is ranges from 20000-73000 µM (20-73 mM)

in comparison to the standard inhibitor these vaIues are very high. Other 36 drugs

showed moderate to good inhibitions with as low as lC50=272 µM (DB-9). Remining

three drugs DB-22, DB-56 and DB-79 were the standard inhibitors of ACE and

they have lC50 vaIue 2.663 µM, 10.026 µM and 8.125 µM, respectiveIy. List of top

10 most active drugs aIong with their aIready used purposes is given beIow in

Table 5.3.

Table 5.3: Ten most active drugs along with their purposes.

Code Name Purpose of Drug lC50 (μM)

DB-009 CiprofIoxacin HCI monohydrate Antiobiotic 272

DB-001 AmIodipine BesyIate CaIcium channeI bIocker, CVS

288

DB-017 ltopride HydrochIoride AChE lnhibitor 296

DB-003 AmpiciIIin Trihydrate Antiobiotic 319

DB-054 DoxycycIine HycIate Antiobiotic 329

DB-053 Diphenhydramine HydrochIoride antihistamine mainIy used to treat aIIergies

338

DB-046 ChIoroquine Phosphate antimaIariaI 342

DB-002 AmoxiciIIin trihydrate Antiobiotic 364

DB-012 DiItiazem hydrochIoride CaIcium channeI bIocker, CVS

490

DB-123 SertraIine HydrochIoride anxity, depression, panic attacks

581

5.3.2 Inhibition Study of Synthetic Compounds

Synthetic compounds were included the derivatives of tryptamine and indole.

Tryptamine is a monoamine alkaloid. It contains an indole ring structure and is

similar to the amino acid tryptophan. While indole is an aromatic heterocyclic

Page 170: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

151

compound which is synthesized by the fusion of a benzene ring with pyrrole ring.

Structures of both compounds are shown below in Table 5.4 A and B.

Four different cIasses of above compounds were used for screening incIuding

“Schiff bases of tryptamine” which were synthesized by treating tryptamine with

different substituted benzaIdehydes in methanoI. The second cIass of “Urea and

thiourea derivatives of tryptamine” were prepared by treating tryptamine with

dichIoromethane and triethyIamine then different substituted isocyanates or

isothiocyanates were added to the reaction mixture. The third cIass of “IndoIe

acryIonitriIes derivatives” were prepared by reacting cyanoacetic acid and indoIe

aIong with acetic anhydride to produce cyanoacetyI indoIe which then react with

corresponding benzaIdehydes in presence of ethanoI and cataIytic amount of

trimethyIamine. The fourth cIass of “Bis(indoIyI) methanes” were synthesized by

reaction of indoIe with various aryI substituted aIdehydes in the presence of grape

juice which was used as cataIyst. GeneraI structures of four cIasses of derivatives

are shown beIow in Table 5.4 C-F.

Table 5.4: Structures of base compounds and general structures of derivatives classes.

A) Tryptamine

B) IndoIe

C) Schiff bases of tryptamine

D) Urea and thiourea derivatives of tryptamine

NH

HN

HN

X

R

Page 171: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

152

E) IndoIe acryIonitriIes

F) Bis(indoIyI) methanes

All compounds of different chemical class and functional groups were prepared in-

house and checked for their inhibitory potential, for this purpose 40 different

compounds were chosen randomly among many other compounds. All

compounds as given in Table 5.5 were prepared and diluted in DMSO and

analysed with standard method.

Page 172: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

153

Table 5.5: Synthetic compounds used for inhibition study.

S.No. Code /

na me

MoIecuIar

formuIa

MoIecuIar

weight

Stru cture Chemical cIass Solvent

us ed

1 KHA-I-35 C17H14Cl2N2O 333

Tryptamine Schiff Bases DMSO

2 KHA-I-39 C18H17BrN2O2 373

Tryptamine Schiff Bases DMSO

Page 173: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

154

3 KHA-I-40 C17H15ClN2O 298

Tryptamine Schiff Bases DMSO

4 KHA-I-51 C18H18N2S 294

Tryptamine Schiff Bases DMSO

5 KHA-I-53 C19H20N2O2 308

Tryptamine Schiff Bases DMSO

Page 174: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

155

6 KHA-I-58 C18H18N2O2 294

Tryptamine Schiff Bases DMSO

7 KHA-I-90 C17H15N3O3 309

Tryptamine Schiff Bases DMSO

8 KHA-II-8 C17H15BrN2O 342

NH

N

H

OH

Br

Tryptamine Schiff Bases DMSO

Page 175: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

156

9 KHA-I-69 C17H17N3S 295

Urea and Thiourea

derivatives

DMSO

10 KHA-I-71 C17H16N4O3 324

Urea and Thiourea

derivatives

DMSO

11 KHA-I-76 C18H16F3N3O 347

Urea and Thiourea

derivatives

DMSO

12 KHA-II-29 C17H16BrN3S 374

Urea and Thiourea

derivatives

DMSO

Page 176: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

157

13 KHA-II-43 C17H16BrN3S 374

Urea and Thiourea

derivatives

DMSO

14 KHA-II-69 C18H19N3OS 325

Urea and Thiourea

derivatives

DMSO

15 KHA-II-73 C19H21N3S 323

Urea and Thiourea

derivatives

DMSO

16 KHA-II-76 C17H16BrN3S 374

Urea and Thiourea

derivatives

DMSO

Page 177: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

158

17 KHA-I-100 C21H18N2O3 346

Indole acrylonitriles DMSO

18 KHA-II-19 C18H11N3O4 333

Indole acrylonitriles DMSO

19 KHA-II-38 C20H16N2O3 332

Indole acrylonitriles DMSO

Page 178: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

159

20 KHA-II-39 C20H15BrN2O3 410

Indole acrylonitriles DMSO

21 KHA-II-51 C22H19BrN2O3 438

Indole acrylonitriles DMSO

22 KHA-II-56 C19H14N2O2 302

Indole acrylonitriles DMSO

Page 179: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

160

23 KHA-II-61 C20H14BrFN2O 396

Indole acrylonitriles DMSO

24 KHA-III-6 C20H16N2O 300

Indole acrylonitriles DMSO

25 KHA-III-10 C24H18N2O 350

Indole acrylonitriles DMSO

Page 180: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

161

26 KHA-IV-39 C22H19BrN2O3 438

Indole acrylonitriles DMSO

27 KHA-III-59 C23H18N2 322

KHA Bis indole DMSO

28 KHA-III-66 C24H20N2O2 368

KHA Bis indole DMSO

Page 181: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

162

29 KHA-III-71 C24H19IN2O2 494

KHA Bis indole DMSO

30 KHA-III-78 C23H17N3O2 367

KHA Bis indole DMSO

31 KHA-III-80 C23H17BrN2 400

KHA Bis indole DMSO

Page 182: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

163

32 KHA-III-90 C23H17BrN2O 416

KHA Bis indole DMSO

33 KHA-III-97 C23H17ClN2 356

KHA Bis indole DMSO

34 KHA-IV-7 C24H20N2 336

KHA Bis indole DMSO

35 KHA-IV-45 C29H30N2O2 438

KHA Bis indole DMSO

Page 183: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

164

36 KHA-IV-47 C29H29BrN2O2 516

KHA Bis indole DMSO

37 KHA-IV-55 C27H25BrN2 456

KHA Bis indole DMSO

38 KHA-IV-62 C29H30N2O2 438

KHA Bis indole DMSO

Page 184: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

165

39 KHA-IV-72 C28H27FN2O 426

KHA Bis indole DMSO

40 KHA-IV-81 C25H24N2S 384

KHA Bis indole DMSO

Page 185: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

166

Total 40 compounds were selected for screening, 8 from tryptamine schiff bases,

8 from urea and thiourea derivatives, 10 from indole acrylonitriles and 14 from KHA

bis indole. All compounds were prepared of a high concentration of 50 mM in

DMSO, those compounds which showed the inhibition were again analyzed with

the low concentration. IC50 value of all compounds is given in Table 5.6.

Table 5.6: Screening results of synthetic compounds, IC50 of active compounds.

S. No. Code / Name IC50 (µM)

1 KHA-I-35 4395

2 KHA-I-39 Inactive

3 KHA-I-40 Inactive

4 KHA-I-51 2936

5 KHA-I-53 933

6 KHA-I-58 441

7 KHA-I-90 522

8 KHA-II-8 1442

9 KHA-I-69 3439

10 KHA-I-71 Inactive

11 KHA-I-76 2116

12 KHA-II-29 1334

13 KHA-II-43 Inactive

14 KHA-II-69 1030

15 KHA-II-73 Inactive

16 KHA-II-76 1703

17 KHA-I-100 673

18 KHA-II-19 5123

19 KHA-II-38 782

20 KHA-II-39 737

21 KHA-II-51 622

22 KHA-II-56 566

Page 186: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

167

23 KHA-II-61 ~50000

24 KHA-III-6 1207

25 KHA-III-10 2785

26 KHA-IV-39 Inactive

27 KHA-III-59 1336

28 KHA-III-66 859

29 KHA-III-71 353

30 KHA-III-78 534

31 KHA-III-80 643

32 KHA-III-90 320

33 KHA-III-97 445

34 KHA-IV-7 990

35 KHA-IV-45 1259

36 KHA-IV-47 1340

37 KHA-IV-55 1449

38 KHA-IV-62 Inactive

39 KHA-IV-72 1397

40 KHA-IV-81 2031

Among these compounds 7 were inactive, 1 compound KHA-II-61 showed very

low inhibition while all other 32 compounds showed significant inhibition activity

ranged from IC50 = 320-5123 µM

5.3.3 Inhibition Study of Natural Compounds and Extracts

Many natural compounds have already been discovered as ACE inhibitors infact

the first inhibitor of ACE was from the snake venom as already discussed in the

introduction of chapter 1. This developed method was also discovered for its

potential for screening of natural products and extracts of plants. For this purpose,

4 natural compounds (Table 5.7) and 3 methanolic extract of plants (Table 5.8),

were investigated for their potential against ACE.

Page 187: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

168

Table 5.7: Natural compounds used for inhibition study.

S.No. Code Name Mοlecular formula

Mοlecular weight

Structure Sοlvent used

1 SC-1 7-Hydroxy Vasicine (Vasicinol)

C11H12N2O2 204.229

Water

2 SC-2 Vasicine C11H12N2O 188.0950

Methanol

3 SC-3 Quercetin C15H10O7 302.24

Methanol

4 SC-4 Vasicinone C11H10N2O2 202.0742

Methanol

Page 188: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

169

Table 5.8: Natural extracts of plants used for inhibition study.

S.No. Code Extract name Name of plant Extract type Solvent used

5 SC-5 Piper longum MeOH

defatted (in 1% MeOH)

Long pepper or

Piper longum

Extract

defatted with

hexane

Methanol

6 SC-6 Fabago MeOH Zygophyllum

fabago Methanolic Methanol

7 SC-7 AVU-M Artemisia

vulgaris Methanolic Methanol

Among total 7 samples, 4 were natural compounds purified in-house and 3 were

natural extracts of plants also extracted in-house.

Natural compounds used in this study are the purified products of Adhatoda vasica

or Justicia adhatoda (SC-1 to 4). It is a medicinaI important pIant and it beIοngs to

ithe famiIy iAcanthaceae (Aslam et al., 2013). It iis a icοmmon imedicinaI pIant iwhich

is widely iused iin iUnani iand iAyurvedic traditiοnaI imedicines ifor irespiratοry itract

idisοrders ilike icοugh, iasthma, irheumatism, iexpectοrant iand ichrοnic ibronchitis

(Singh et al., 2017).

Lοng ipepper or iPiper Iοngum (SC-5) sοmetimes caIIed as lndian Iοng pepper or

pipli, is a fIοwering ivine and belοngs to ithe ifamiIy iPiperaceae. It is cultivated ifοr iits

ifruit, iwhich iis generally idried iand iused ias ispice. It also iused iin ithe itreatment οf

irespiratοry and idigestive disοrders.

Zygophyllum fabago (SC-6) is commonly iknown ias iSyrian ibean-caper. iIt iis

iconsidered ias a inοxious iweed of ieconοmic iimpοrtance in imany regions iof the

iwestern iUnited States. iIt iis inative to iAsia iand iEurοpe.

Artemisia vulgaris (SC-7) commonly know as mugwort is mostly found in

temperate Europe, Asia and northern Africa, where it is considered as invasive

weed. It is used for pain relif, treamt of fever and it is also used as diuretic agent.

Compounds 1-4 were prepared in a high concentration of 50 mM and extracts 5-

7 were prepared in high concentration of 5000 µg/mL (ppm).

Page 189: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

170

Table 5.9: Results of natural compounds and extracts, IC50 of active samples.

S.No. Code Name IC50 Value

1 SC-1 7-Hydroxy Vasicine 9705 µM

2 SC-2 Vasicine 496 µM

3 SC-3 Quercetin 7124 µM

4 SC-4 Vasicinone 2990 µM

5 SC-5 Piper longum Inactive

6 SC-6 Zygophyllum fabago Inactive

7 SC-7 Artemisia vulgaris 10779 µg/mL

(ppm)

All four natural compounds showed moderate activity, with IC50 value 496 µM -

9705 µM; while in plant extracts only SC-7 of “Artemisia vulgaris” showed some

inhibitory activity with IC50 of 10779 µg/mL (ppm) while other did not showed any

activity. Results are tabulated in Table 5.9.

5.4 Conclusion

Developed method of MALDI-MS had proved to be very effective for screening of

different types of compounds or drugs. In this work, a total of 124 different

compounds/samples from different classes of compounds, including 77

commercial drugs, 40 synthetic compounds, 4 natural products and 3 extracts of

plants were investigated for their inhibitory potential. Among these compounds /

samples a range of activity was founds in different compounds.

In commercial drugs, 36 drugs showed moderate to good inhibition of as low as

IC50 = 272 µM. In drugs analysis, 3 ACE standard inhibitors were also screened

blind folded, which further proved the authenticity and reproducibility of the assay.

Those drugs which found to be active against ACE can be further studied for their

repurposing as ACE inhibitors. In synthetic compounds, 32 compounds showed

significant inhibition ranged from IC50 = 320-5123 µM. Further derivatives of those

Page 190: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

171

drugs and standard inhibitors of ACE can be synthesized for “structural activity

relationship study”, to find more potent inhibitors of ACE. In the last category, all 4

natural compounds were moderately active with IC50 = 496-9705 µM; while only

one plant extracts showed some inhibition with IC50 = 10779 µg/mL (ppm).

Among screened compounds most οf them were found to be good to moderate

inhibition activity against ACE. This methodology ifurther ican ibe iused ifοr ithe

iscreening iοf different drug libraries itο iidentify the Iead icοmpounds as more potent

inhibitors of ACE.

Page 191: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

172

References

Abu-Reidah, I. M., Ali-Shtayeh, M. S., Jamous, R. M., Arráez-Román, D., and Segura-Carretero, A. (2015). HPLC–DAD–ESI-MS/MS screening of bioactive components from Rhus coriaria L. (Sumac) fruits. Food Chemistry, 166, 179-191.

Allen, F., Greiner, R., and Wishart, D. (2015). Competitive fragmentation modeling of ESI-MS/MS spectra for putative metabolite identification. Metabolomics, 11(1), 98-110.

Alves, M., Araujo, M. d. C., Juliano, M. A., Oliveira, E. M. d., Krieger, J. E., Casarini, D. E., Juliano, L., and Carmona, A. K. (2005). A continuous fluorescent assay for the determination of plasma and tissue angiotensin I-converting enzyme activity. Brazilian Journal of Medical and Biological Research, 38(6), 861-868.

Annis, D. A., Athanasopoulos, J., Curran, P. J., Felsch, J. S., Kalghatgi, K., Lee, W. H., Nash, H. M., Orminati, J.-P. A., Rosner, K. E., Shipps, G. W., Thaddupathy, G. R. A., Tyler, A. N., Vilenchik, L., Wagner, C. R., and Wintner, E. A. (2004). An affinity selection–mass spectrometry method for the identification of small molecule ligands from self-encoded combinatorial libraries: Discovery of a novel antagonist of E. coli dihydrofolate reductase. International Journal of Mass Spectrometry, 238(2), 77-83.

Araujo, M. C., Melo, R. I., Del Nery, E., Alves, M. F., Juliano, M. A., Casarini, D. E., Juliano, L., and Carmona, A. K. (1999). Internally quenched fluorogenic substrates for angiotensin I-converting enzyme. Journal of Hypertension, 17(5), 665-672.

Aslam, M., Rais, S., Alam, M., and Pugazhendi, A. (2013). Adsorption of Hg (II) from aqueous solution using Adulsa (Justicia adhatoda) leaves powder: Kinetic and equilibrium studies. Journal of Chemistry, 2013.

Baca, M., and Kent, S. B. H. (1992). Direct observation of a ternary complex between the dimeric enzyme HIV-1 protease and a substrate-based inhibitor. Journal of the American Chemical Society, 114(10), 3992-3993.

Bae, Y. J., Park, K. M., and Kim, M. S. (2012). Reproducibility of temperature-selected mass spectra in matrix-assisted laser desorption ionization of peptides. Analytical Chemistry, 84(16), 7107-7111.

Bakhle, Y. S. (1968). Conversion of angiotensin I to angiotensin II by cell-free extracts of dog lung. Nature, 220(5170), 919-921.

Bakhle, Y. S., Reynard, A. M., and Vane, J. R. (1969). Metabolism of the angiotensins in isolated perfused tissues. Nature, 222(5197), 956-959.

Balasuriya, B. N., and Rupasinghe, H. V. (2011). Plant flavonoids as angiotensin converting enzyme inhibitors in regulation of hypertension. Functional Foods in Health and Disease, 1(5), 172-188.

Page 192: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

173

Balbaa, M., and El Ashry, E. (2012). Enzyme inhibitors as therapeutic tools. Biochemistry & Physiology: Open Access, 1, 1-8.

Barbosa-Filho, J. M., Martins, V. K. M., Rabelo, L. A., Moura, M. D., Silva, M. S., Cunha, E. V. L., Souza, M. F. V., Almeida, R. N., and Medeiros, I. A. (2006). Natural products inhibitors of the angiotensin converting enzyme (ACE): A review between 1980 - 2000. Revista Brasileira de Farmacognosia, 16, 421-446.

Beavis, R. C., Chaudhary, T., and Chait, B. T. (1992). α-Cyano-4-hydroxycinnamic acid as a matrix for matrix assisted laser desorption mass spectromtry. Organic Mass Spectrometry, 27(2), 156-158.

Brenda-Enzyme. (2019). EC 3.4.15.1 - peptidyl-dipeptidase A. Retrieved from https://www.brenda-enzymes.org/enzyme.php?ecno=3.4.15.1

Brodbelt, J. S., and Wilson, J. J. (2009). Infrared multiphoton dissociation in quadrupole ion traps. Mass Spectrometry Reviews, 28(3), 390-424.

Bu, H.-Z., Magis, L., Knuth, K., and Teitelbaum, P. (2001). High-throughput cytochrome P450 (CYP) inhibition screening via a cassette probe-dosing strategy. VI. Simultaneous evaluation of inhibition potential of drugs on human hepatic isozymes CYP2A6, 3A4, 2C9, 2D6 and 2E1. Rapid Communications in Mass Spectrometry, 15(10), 741-748.

Cancilla, M. T., Leavell, M. D., Chow, J., and Leary, J. A. (2000). Mass spectrometry and immobilized enzymes for the screening of inhibitor libraries. Proceedings of the National Academy of Sciences, 97(22), 12008-12013.

Carey, R. M. (2015). The intrarenal renin-angiotensin system in hypertension. Advances in Chronic Kidney Disease, 22(3), 204-210.

Cer, R. Z., Mudunuri, U., Stephens, R., and Lebeda, F. J. (2009). IC50-to-Ki: a web-based tool for converting IC50 to Ki values for inhibitors of enzyme activity and ligand binding. Nucleic Acids Research, 37(suppl_2), W441-W445.

Chan, N. W. C., Lewis, D. F., Rosner, P. J., Kelly, M. A., and Schriemer, D. C. (2003). Frontal affinity chromatography–mass spectrometry assay technology for multiple stages of drug discovery: applications of a chromatographic biosensor. Analytical Biochemistry, 319(1), 1-12.

Chen, J., Wang, Y., Ye, R., Wu, Y., and Xia, W. (2013). Comparison of analytical methods to assay inhibitors of angiotensin I-converting enzyme. Food Chemistry, 141(4), 3329-3334.

Cheung, H.-S., Wang, F.-L., Ondetti, M. A., Sabo, E. F., and Cushman, D. W. (1980). Binding of peptide substrates and inhibitors of angiotensin-converting enzyme. Importance of the COOH-terminal dipeptide sequence. Journal of Biological Chemistry, 255(2), 401-407.

Page 193: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

174

Chu, I., Favreau, L., Soares, T., Lin, C.-C., and Nomeir, A. A. (2000). Validation of higher-throughput high-performance liquid chromatography/atmospheric pressure chemical ionization tandem mass spectrometry assays to conduct cytochrome P450s CYP2D6 and CYP3A4 enzyme inhibition studies in human liver microsomes. Rapid Communications in Mass Spectrometry, 14(4), 207-214.

Cífková, E., Holčapek, M., Lísa, M., Vrána, D., Gatěk, J., and Melichar, B. (2015). Determination of lipidomic differences between human breast cancer and surrounding normal tissues using HILIC-HPLC/ESI-MS and multivariate data analysis. Analytical and Bioanalytical Chemistry, 407(3), 991-1002.

Comley, J. (2009). Design of experiments. Drug Discovery, 10, 41.

Cui, L., Nithipatikom, K., and Campbell, W. B. (2007). Simultaneous analysis of angiotensin peptides by LC–MS and LC–MS/MS: Metabolism by bovine adrenal endothelial cells. Analytical Biochemistry, 369(1), 27-33.

Davis, R. G., Anderegg, R. J., and Blanchard, S. G. (1999). Iterative size-exclusion chromatography coupled with liquid chromatographic mass spectrometry to enrich and identify tight-binding ligands from complex mixtures. Tetrahedron, 55(39), 11653-11667.

de Boer, A. R., Lingeman, H., Niessen, W. M., and Irth, H. (2007). Mass spectrometry-based biochemical assays for enzyme-inhibitor screening. Trac Trends in Analytical Chemistry, 26(9), 867-883.

de Rond, T., Danielewicz, M., and Northen, T. (2015). High throughput screening of enzyme activity with mass spectrometry imaging. Current Opinion in Biotechnology, 31, 1-9.

Deng, G., Gu, R.-F., Marmor, S., Fisher, S. L., Jahic, H., and Sanyal, G. (2004). Development of an LC–MS based enzyme activity assay for MurC: application to evaluation of inhibitors and kinetic analysis. Journal of Pharmaceutical and Biomedical Analysis, 35(4), 817-828.

Dunayevskiy, Y. M., Lai, J.-J., Quinn, C., Talley, F., and Vouros, P. (1997). Mass spectrometric identification of ligands selected from combinatorial libraries using gel filtration. Rapid Communications in Mass Spectrometry, 11(11), 1178-1184.

Dunbar, R. C. (2004). BIRD (blackbody infrared radiative dissociation): Evolution, principles, and applications. Mass Spectrometry Reviews, 23(2), 127-158.

El-Dorry, H., Bull, H. G., Iwata, K., Thornberry, N. A., Cordes, E. H., and Soffer, R. (1982). Molecular and catalytic properties of rabbit testicular dipeptidyl carboxypeptidase. Journal of Biological Chemistry, 257(23), 14128-14133.

Elased, K. M., Cunha, T. S., Gurley, S. B., Coffman, T. M., and Morris, M. (2006). New mass spectrometric assay for angiotensin-converting enzyme 2 activity. Hypertension, 47(5), 1010-1017.

Page 194: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

175

ESRF. Human angiotensin I converting enzyme structure revealed. Retrieved from http://www.esrf.eu/UsersAndScience/Publications/Highlights/2003/MX/MX10

Ferreira, S. (1965). A bradykinin‐potentiating factor (BPF) present in the venom of Bothrops jararaca. British Journal of Pharmacology and Chemotherapy, 24(1), 163-169.

Ferreira, S. H., Bartelt, D. C., and Greene, L. J. (1970). Isolation of bradykinin-potentiating peptides from Bothrops jararaca venom. Biochemistry, 9(13), 2583-2593.

Fu, Z., Yun, S. Y., Won, J. H., Back, M. J., Jang, J. M., Ha, H. C., Lee, H. K., Shin, I. C., Kim, J. Y., Kim, H. S., and Kim, D. K. (2019). Development of a label-free LC-MS/MS-based glucosylceramide synthase assay and its application to inhibitors screening for ceramide-related diseases. Biomolecules & Therapeutics, 27(2), 193-200.

Gao, H., and Leary, J. A. (2003). Multiplex inhibitor screening and kinetic constant determinations for yeast hexokinase using mass spectrometry based assays. Journal of the American Society for Mass Spectrometry, 14(3), 173-181.

Gao, J., Cheng, X., Chen, R., Sigal, G. B., Bruce, J. E., Schwartz, B. L., Hofstadler, S. A., Anderson, G. A., Smith, R. D., and Whitesides, G. M. (1996). Screening derivatized peptide libraries for tight binding inhibitors to carbonic anhydrase II by electrospray ionization-mass spectrometry. Journal of Medicinal Chemistry, 39(10), 1949-1955.

Greco, G., Boltner, A., and Letzel, T. (2014). Optimization of jet stream ESI parameters when coupling Agilent 1260 infinity analytical SFC system with Agilent 6230 TOF-LC/MS.

Greis, K. D. (2007). Mass spectrometry for enzyme assays and inhibitor screening: an emerging application in pharmaceutical research. Mass Spectrometry Reviews, 26(3), 324-339.

Greis, K. D., Zhou, S., Burt, T. M., Carr, A. N., Dolan, E., Easwaran, V., Evdokimov, A., Kawamoto, R., Roesgen, J., and Davis, G. F. (2006). MALDI-TOF MS as a label-free approach to rapid inhibitor screening. Journal of the American Society for Mass Spectrometry, 17(6), 815-822.

Guerrero, L., Castillo, J., Quiñones, M., Garcia-Vallvé, S., Arola, L., Pujadas, G., and Muguerza, B. (2012). Inhibition of angiotensin-converting enzyme activity by flavonoids: Structure-activity relationship studies. PloS One, 7(11), e49493-e49493.

Page 195: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

176

Henda, Y. B., Labidi, A., Arnaudin, I., Bridiau, N., Delatouche, R., Maugard, T., Piot, J.-M., Sannier, F., Thiéry, V., and Bordenave-Juchereau, S. (2013). Measuring angiotensin-i converting enzyme inhibitory activity by micro plate assays: Comparison using marine cryptides and tentative threshold determinations with captopril and losartan. Journal of Agricultural and Food Chemistry, 61(45), 10685-10690.

Hodgson, R. J., Besanger, T. R., Brook, M. A., and Brennan, J. D. (2005). Inhibitor screening using immobilized enzyme reactor chromatography/mass spectrometry. Analytical Chemistry, 77(23), 7512-7519.

Holle, A., Haase, A., Kayser, M., and Höhndorf, J. (2006). Optimizing UV laser focus profiles for improved MALDI performance. Journal of Mass Spectrometry, 41(6), 705-716.

Holle, A., and Ketterlinus, R. (2008). Innovative smartbeam laser technology enhances MALDI-TOF based proteomics applications. Technical Notes.

Holmquist, B., Bünning, P., and Riordan, J. F. (1979). A continuous spectrophotometric assay for angiotensin converting enzyme. Analytical Biochemistry, 95(2), 540-548.

Hooda, A., Nanda, A., Jain, M., Kumar, V., and Rathee, P. (2012). Optimization and evaluation of gastroretentive ranitidine HCl microspheres by using design expert software. International Journal of Biological Macromolecules, 51(5), 691-700.

Hsieh, F., Keshishian, H., and Muir, C. (1998). Automated high throughput multiple target screening of molecular libraries by microfluidic MALDI-TOF MS. Journal of Biomolecular Screening, 3(3), 189-198.

Ingkaninan, K., de Best, C. M., van der Heijden, R., Hofte, A. J. P., Karabatak, B., Irth, H., Tjaden, U. R., van der Greef, J., and Verpoorte, R. (2000). High-performance liquid chromatography with on-line coupled UV, mass spectrometric and biochemical detection for identification of acetylcholinesterase inhibitors from natural products. Journal of Chromatography A, 872(1), 61-73.

Ingkaninan, K., Hazekamp, A., de Best, C. M., Irth, H., Tjaden, U. R., van der Heijden, R., van der Greef, J., and Verpoorte, R. (2000). The application of HPLC with on-line coupled UV/MS−biochemical detection for isolation of an acetylcholinesterase inhibitor from narcissus ‘Sir Winston Churchill'. Journal of Natural Products, 63(6), 803-806.

IPFS. Electrospray ionization. Retrieved from https://ipfs.io/ipfs/QmXoypizjW3WknFiJnKLwHCnL72vedxjQkDDP1mXWo6uco/wiki/Electrospray_ionization.html

Jackson, M. (2019). Triple quadrupole schematic.jpeg. Retrieved from https://en.wikipedia.org/wiki/File:Triple_quadrupole_schematic.jpeg

Page 196: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

177

Johnson, B. M., Nikolic, D., and van Breemen, R. B. (2002). Applications of pulsed ultrafiltration-mass spectrometry. Mass Spectrometry Reviews, 21(2), 76-86.

Kang, D. G., Yun, Y. G., Ryoo, J. H., and Lee, H. S. (2002). Anti-hypertensive effect of water extract of danshen on renovascular hypertension through inhibition of the renin angiotensin system. The American Journal of Chinese Medicine, 30(01), 87-93.

Karra, V. K., Mullangi, R., Pilli, N. R., Inamadugu, J. K., Ravi, V. B., and Seshagiri Rao, J. (2012). A rapid and sensitive liquid chromatography–tandem mass spectrometric assay for moexipril, an angiotensin‐converting enzyme inhibitor in human plasma. Biomedical Chromatography, 26(12), 1552-1558.

Kenakin, T. P. (2019). Chapter 11 - The Drug Discovery Process. In T. P. Kenakin (Ed.), A Pharmacology Primer (Fifth Edition) (pp. 323-371): Academic Press.

Khan, O. (2019). Mass analyzers (mass spectrometry). Retrieved from https://chem.libretexts.org/Bookshelves/Analytical_Chemistry/Supplemental_Modules_(Analytical_Chemistry)/Instrumental_Analysis/Mass_Spectrometry/Mass_Spectrometers_(Instrumentation)/Mass_Analyzers_(Mass_Spectrometry)

Lahogue, V., Réhel, K., Taupin, L., Haras, D., and Allaume, P. (2010). A HPLC-UV method for the determination of angiotensin I-converting enzyme (ACE) inhibitory activity. Food Chemistry, 118(3), 870-875.

Lapointe, N., and Rouleau, J.-L. (2002). Activation of vascular tissue angiotensin-converting enzyme (ACE) in heart failure: Effects of ACE inhibitors. Journal of the American College of Cardiology, 39(5), 776-779.

Lee, C., Lu, I. C., Hsu, H. C., Lin, H.-Y., Liang, S.-P., Lee, Y.-T., and Ni, C.-K. (2016). Formation of metal-related ions in matrix-assisted laser desorption ionization. Journal of the American Society for Mass Spectrometry, 27(9), 1491-1498.

Li, G.-H., Liu, H., Shi, Y.-H., and Le, G.-W. (2005). Direct spectrophotometric measurement of angiotensin I-converting enzyme inhibitory activity for screening bioactive peptides. Journal of Pharmaceutical and Biomedical Analysis, 37(2), 219-224.

Liesener, A., and Karst, U. (2005). Monitoring enzymatic conversions by mass spectrometry: a critical review. Analytical and Bioanalytical Chemistry, 382(7), 1451-1464.

Liesener, A., Perchuc, A. M., Schöni, R., Wilmer, M., and Karst, U. (2005). Screening for proteolytic activities in snake venom by means of a multiplexing electrospray ionization mass spectrometry assay scheme. Rapid Communications in Mass Spectrometry, 19(20), 2923-2928.

Page 197: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

178

Lu, C.-Y., Liu, F.-T., and Feng, C.-H. (2011). Quantitation of quinapril in human plasma by matrix-assisted laser desorption ionization time-of-flight mass spectrometry with quinolone matrix additives. Journal of Chromatography B, 879(26), 2688-2694.

Luque de Castro, M. D. (2019). Flow analysis | flow injection analysis: Instrumentation. In P. Worsfold, C. Poole, A. Townshend, and M. Miró (Eds.), Encyclopedia of Analytical Science (Third Edition) (pp. 204-212). Oxford: Academic Press.

Lyubarskaya, Y. V., Carr, S. A., Dunnington, D., Prichett, W. P., Fisher, S. M., Appelbaum, E. R., Jones, C. S., and Karger, B. L. (1998). Screening for high-affinity ligands to the Src SH2 domain using capillary isoelectric focusing-electrospray ionization ion trap mass spectrometry. Analytical Chemistry, 70(22), 4761-4770.

Martin, M., and Deussen, A. (2019). Effects of natural peptides from food proteins on angiotensin converting enzyme activity and hypertension. Critical Reviews in Food Science and Nutrition, 59(8), 1264-1283.

Mason, D. R., Reid, J. D., Camenzind, A. G., Holmes, D. T., and Borchers, C. H. (2012). Duplexed iMALDI for the detection of angiotensin I and angiotensin II. Methods, 56(2), 213-222.

Mathur, S., Park, J. D., Kim, D. H., and Hartmann, R. W. (2005). A method for screening enzyme inhibitors using size exclusion chromatography and ESI-LC-MS/MS. Journal of Biomolecular Screening, 10(1), 30-35.

MEMBERS, W. G., Benjamin, E. J., Blaha, M. J., Chiuve, S. E., Cushman, M., Das, S. R., Deo, R., de Ferranti, S. D., Floyd, J., and Fornage, M. (2017). Heart disease and stroke statistics—2017 update: a report from the American Heart Association. Circulation, 135(10), e146.

Menard, J., and Patchett, A. A. (2001). Angiotensin-converting enzyme inhibitors. Advances in Protein Chemistry, 56, 13-75.

Min, W., Cui, S., Wang, W., Chen, J., and Hu, Z. (2013). Capillary electrophoresis applied to screening of trypsin inhibitors using microreactor with trypsin immobilized by glutaraldehyde. Analytical Biochemistry, 438(1), 32-38.

Mirzaian, M., Wisse, P., Ferraz, M. J., Marques, A. R. A., Gaspar, P., Oussoren, S. V., Kytidou, K., Codée, J. D. C., van der Marel, G., Overkleeft, H. S., and Aerts, J. M. (2017). Simultaneous quantitation of sphingoid bases by UPLC-ESI-MS/MS with identical 13C-encoded internal standards. Clinica Chimica Acta, 466, 178-184.

Mistarz, U. H., Bellina, B., Jensen, P. F., Brown, J. M., Barran, P. E., and Rand, K. D. (2018). UV photodissociation mass spectrometry accurately localize sites of backbone deuteration in peptides. Analytical Chemistry, 90(2), 1077-1080.

Page 198: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

179

Mordehai, A., and Fjeldsted, J. (2009). Agilent jet stream thermal gradient focusing technology. Agilent Technologies Technical Note, Publication Number 5990, 3494.

Moy, F. J., Haraki, K., Mobilio, D., Walker, G., Powers, R., Tabei, K., Tong, H., and Siegel, M. M. (2001). MS/NMR:  A structure-based approach for discovering protein ligands and for drug design by coupling size exclusion chromatography, mass spectrometry, and nuclear magnetic resonance spectroscopy. Analytical Chemistry, 73(3), 571-581.

Muckenschnabel, I., Falchetto, R., Mayr, L. M., and Filipuzzi, I. (2004). SpeedScreen: label-free liquid chromatography–mass spectrometry-based high-throughput screening for the discovery of orphan protein ligands. Analytical Biochemistry, 324(2), 241-249.

Musharraf, S. G., Bhatti, M. S., Choudhary, M. I., and Rahman, A.-u. (2017). Screening of inhibitors of angiotensin-converting enzyme (ACE) employing high performance liquid chromatography-electrospray ionization triple quadrupole mass spectrometry (HPLC-ESI-QqQ-MS). European Journal of Pharmaceutical Sciences, 101, 182-188.

Natesh, R., Schwager, S. L., Sturrock, E. D., and Acharya, K. R. (2003). Crystal structure of the human angiotensin-converting enzyme–lisinopril complex. Nature, 421(6922), 551.

Nikolic, D., Habibi-Goudarzi, S., Corley, D. G., Gafner, S., Pezzuto, J. M., and van Breemen, R. B. (2000). Evaluation of cyclooxygenase-2 inhibitors using pulsed ultrafiltration mass spectrometry. Analytical Chemistry, 72(16), 3853-3859.

Norris, A. J., Whitelegge, J. P., Faull, K. F., and Toyokuni, T. (2001). Analysis of enzyme kinetics using electrospray ionization mass spectrometry and multiple reaction monitoring:  Fucosyltransferase V. Biochemistry, 40(13), 3774-3779.

Ortiz-Salmerón, E., Barón, C., and Garcıa-Fuentes, L. (1998). Enthalpy of captopril‐angiotensin I‐converting enzyme binding. FEBS Letters, 435(2-3), 219-224.

Özbal, C. C., LaMarr, W. A., Linton, J. R., Green, D. F., Katz, A., Morrison, T. B., and Brenan, C. J. H. (2004). High throughput screening via mass spectrometry: A case study using acetylcholinesterase. ASSAY and Drug Development Technologies, 2(4), 373-382.

Palmer, T., and Bonner, P. L. (2007). Enzymes: biochemistry, biotechnology, clinical chemistry: Elsevier.

Palmer, T., and Bonner, P. L. (2011). 8 - Enzyme Inhibition. In T. Palmer and P. L. Bonner (Eds.), Enzymes (Second Edition) (pp. 126-152): Woodhead Publishing.

Page 199: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

180

Park, K. M., Bae, Y. J., Ahn, S. H., and Kim, M. S. (2012). A simple method for quantification of peptides and proteins by matrix-assisted laser desorption ionization mass spectrometry. Analytical Chemistry, 84(23), 10332-10337.

Percy, A. J., Yang, J., Chambers, A. G., and Borchers, C. H. (2016). Increased depth and breadth of plasma protein quantitation via two-dimensional liquid chromatography/multiple reaction monitoring-mass spectrometry with labeled peptide standards. In S. Sechi (Ed.), Quantitative Proteomics by Mass Spectrometry (pp. 1-21). New York, NY: Springer New York.

Rathore, R., Corr, J., Scott, G., Vollmerhaus, P., and Greis, K. D. (2008). Development of an inhibitor screening platform via mass spectrometry. Journal of Biomolecular Screening, 13(10), 1007-1013.

Rayleigh, L. (1882). XX. On the equilibrium of liquid conducting masses charged with electricity. The London, Edinburgh, and Dublin Philosophical Magazine and Journal of Science, 14(87), 184-186.

Rhee, I. K., Appels, N., Hofte, B., Karabatak, B., Erkelens, C., Stark, L. M., Flippin, L. A., and Verpoorte, R. (2004). Isolation of the acetylcholinesterase inhibitor ungeremine from Nerine bowdenii by preparative HPLC coupled on-line to a flow assay system. Biological and Pharmaceutical Bulletin, 27(11), 1804-1809.

Rockwood, A. L., Kushnir, M. M., and Clarke, N. J. (2018). 2 - Mass Spectrometry. In N. Rifai, A. R. Horvath, and C. T. Wittwer (Eds.), Principles and Applications of Clinical Mass Spectrometry (pp. 33-65): Elsevier.

Ruhaak, L. R. (2017). The use of multiple reaction monitoring on QQQ-MS for the analysis of protein- and site-specific glycosylation patterns in serum. In G. Lauc and M. Wuhrer (Eds.), High-Throughput Glycomics and Glycoproteomics: Methods and Protocols (pp. 63-82). New York, NY: Springer New York.

Sakharov, I. Y., Danilov, S. M., and Dukhanina, E. A. (1987). Affinity chromatography and some properties of the angiotensin-converting enzyme from human heart. Biochimica et Biophysica Acta (BBA) - General Subjects, 923(1), 143-149.

Sannes-Lowery, K. A., Cummins, L. L., Chen, S., Drader, J. J., and Hofstadler, S. A. (2004). High throughput drug discovery with ESI-FTICR. International Journal of Mass Spectrometry, 238(2), 197-206.

Schriemer, D. C. (2004). Peer reviewed: biosensor alternative: frontal affinity chromatography: ACS Publications.

Shalaby, S. M., Zakora, M., and Otte, J. (2006). Performance of two commonly used angiotensin-converting enzyme inhibition assays using FA-PGG and HHL as substrates. Journal of Dairy Research, 73(2), 178-186.

Page 200: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

181

Shi, L., Mao, C., Xu, Z., and Zhang, L. (2010). Angiotensin-converting enzymes and drug discovery in cardiovascular diseases. Drug Discovery Today, 15(9-10), 332-341.

Siegel, M. M., Tabei, K., Bebernitz, G. A., and Baum, E. Z. (1998). Rapid methods for screening low molecular mass compounds non-covalently bound to proteins using size exclusion and mass spectrometry applied to inhibitors of human cytomegalovirus protease. Journal of Mass Spectrometry, 33(3), 264-273.

Silverman, R. B., and Holladay, M. W. (2014). The organic chemistry of drug design and drug action: Academic press.

Singh, A., Kumar, S., Bajpai, V., and Kumar, B. (2017). Simultaneous determination of pyrroquinazoline alkaloids and flavonoids in Adhatoda beddomei and Adhatoda vasica and their marketed herbal formulations using ultra-high-performance liquid chromatography coupled with triple quadrupole linear ion trap mass spectrometry. Biomedical Chromatography, 31(3), e3827.

Sirisha, V. L., Jain, A., and Jain, A. (2016). Enzyme immobilization: An overview on methods, support material, and applications of immobilized enzymes. In S.-K. Kim and F. Toldrá (Eds.), Advances in Food and Nutrition Research (Vol. 79, pp. 179-211): Academic Press.

Sleno, L., and Volmer, D. A. (2004). Ion activation methods for tandem mass spectrometry. Journal of Mass Spectrometry, 39(10), 1091-1112.

Slon-Usakiewicz, J. J., Ng, W., Dai, J.-R., Pasternak, A., and Redden, P. R. (2005). Frontal affinity chromatography with MS detection (FAC-MS) in drug discovery. Drug Discovery Today, 10(6), 409-416.

Slon-Usakiewicz, J. J., Ng, W., Foster, J. E., Dai, J.-R., Deretey, E., Toledo-Sherman, L., Redden, P. R., Pasternak, A., and Reid, N. (2004). Frontal affinity chromatography with MS detection of EphB2 tyrosine kinase receptor. 1. comparison with conventional ELISA. Journal of Medicinal Chemistry, 47(21), 5094-5100.

Smirnov, I. P., Zhu, X., Taylor, T., Huang, Y., Ross, P., Papayanopoulos, I. A., Martin, S. A., and Pappin, D. J. (2004). Suppression of α-cyano-4-hydroxycinnamic acid matrix clusters and reduction of chemical noise in MALDI-TOF mass spectrometry. Analytical Chemistry, 76(10), 2958-2965.

Smolira, A., and Wessely-Szponder, J. (2015). Importance of the matrix and the matrix/sample ratio in MALDI-TOF-MS analysis of cathelicidins obtained from porcine neutrophils. Applied Biochemistry and Biotechnology, 175(4), 2050-2065.

Page 201: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

182

Soderberg, T. (2019). 4.1: Mass Spectrometry. Retrieved from https://chem.libretexts.org/Bookshelves/Organic_Chemistry/Book%3A_Organic_Chemistry_with_a_Biological_Emphasis_(Soderberg)/Chapter_04%3A_Structure_Determination_I/4.1%3A_Mass_Spectrometry

Somogyi, Á. (2008). Chapter 6 - Mass spectrometry instrumentation and techniques. In K. Vékey, A. Telekes, and A. Vertes (Eds.), Medical Applications of Mass Spectrometry (pp. 93-140). Amsterdam: Elsevier.

Song, F., El-Demerdash, A., and Lee, S.-J. S. H. (2012). Screening for multiple phosphodiesterase type 5 inhibitor drugs in dietary supplement materials by flow injection mass spectrometry and their quantification by liquid chromatography tandem mass spectrometry. Journal of Pharmaceutical and Biomedical Analysis, 70, 40-46.

Stevenson, D. E., Feng, R., and Storer, A. C. (1990). Detection of covalent enzyme-substrate complexes of nitrilase by ion-spray mass spectroscopy. FEBS Letters, 277(1-2), 112-114.

Stokvis, E., Clugston, S. L., Honek, J. F., and Heck, A. J. (2000). Characterization of Glyoxalase I (E. Coli)—Inhibitor interactions by electrospray time-of-flight mass spectrometry and enzyme kinetic analysis. Journal of Protein Chemistry, 19(5), 389-397.

Szot, C., and Croxatto, A. (2019). MALDI-TOF diagram. Retrieved from https://en.wikipedia.org/wiki/File:MALDI_TOF_EN.png

Takayama, S., Martin, R., Wu, J., Laslo, K., Siuzdak, G., and Wong, C.-H. (1997). Chemoenzymatic preparation of novel cyclic imine sugars and rapid biological activity evaluation using electrospray mass spectrometry and kinetic analysis. Journal of the American Chemical Society, 119(35), 8146-8151.

The Binding Database. Angiotensin-Converting Enzyme. Retrieved from https://www.bindingdb.org/jsp/dbsearch/PrimarySearch_ki.jsp?target=Angiotensin-converting+enzyme&tag=tg&kiunit=nM&icunit=nM&column=ki&submit=Search&energyterm=kJ%2Fmole

van Breemen, R. B., Huang, C.-R., Nikolic, D., Woodbury, C. P., Zhao, Y.-Z., and Venton, D. L. (1997). Pulsed ultrafiltration mass spectrometry:  A new method for screening combinatorial libraries. Analytical Chemistry, 69(11), 2159-2164.

van Elswijk, D. A., Diefenbach, O., van der Berg, S., Irth, H., Tjaden, U. R., and van der Greef, J. (2003). Rapid detection and identification of angiotensin-converting enzyme inhibitors by on-line liquid chromatography–biochemical detection, coupled to electrospray mass spectrometry. Journal of Chromatography A, 1020(1), 45-58.

Page 202: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

183

Verdugo, D. E., Cancilla, M. T., Ge, X., Gray, N. S., Chang, Y.-T., Schultz, P. G., Negishi, M., Leary, J. A., and Bertozzi, C. R. (2001). Discovery of estrogen sulfotransferase inhibitors from a purine library screen. Journal of Medicinal Chemistry, 44(17), 2683-2686.

Wabnitz, P. A., and Loo, J. A. (2002). Drug screening of pharmaceutical discovery compounds by micro-size exclusion chromatography/mass spectrometry. Rapid Communications in Mass Spectrometry, 16(2), 85-91.

Wang, C., Tian, J., and Wang, Q. (2011). ACE inhibitory and antihypertensive properties of apricot almond meal hydrolysate. European Food Research and Technology, 232(3), 549-556.

Wang, J., Liu, S., Li, S., Song, F., Zhang, Y., Liu, Z., and Liu, C.-m. (2014). Ultrafiltration LC-PDA-ESI/MS combined with reverse phase-medium pressure liquid chromatography for screening and isolation potential α-glucosidase inhibitors from Scutellaria baicalensis Georgi. Analytical Methods, 6(15), 5918-5924.

Werner, C., Pöss, J., and Böhm, M. (2010). Optimal antagonism of the renin-angiotensin-aldosterone system. Drugs, 70(10), 1215-1230.

Wikipedia. Renin angiotensin aldosterone system Retrieved from https://upload.wikimedia.org/wikipedia/commons/3/36/Renin-angiotensin-aldosterone_system.svg

Wong, M. K. (2016). Angiotensin converting enzymes Handbook of Hormones (pp. 263-e229D-264): Elsevier.

Wu, G., Yuan, Y., and Hodge, C. N. (2003). Determining appropriate substrate conversion for enzymatic assays in high-throughput screening. Journal of Biomolecular Screening, 8(6), 694-700.

Wu, J., Aluko, R. E., and Muir, A. D. (2002). Improved method for direct high-performance liquid chromatography assay of angiotensin-converting enzyme-catalyzed reactions. Journal of Chromatography A, 950(1), 125-130.

Wu, J., Takayama, S., Wong, C.-H., and Siuzdak, G. (1997). Quantitative electrospray mass spectrometry for the rapid assay of enzyme inhibitors. Chemistry & Biology, 4(9), 653-657.

Xiao, X., Luo, X., Chen, B., and Yao, S. (2006). Determination of angiotensin converting enzyme inhibitory activity by high-performance liquid chromatography/electrospray-mass spectrometry. Journal of Chromatography B, 834(1), 48-54.

Yamashita, M., and Fenn, J. B. (1984). Electrospray ion source. Another variation on the free-jet theme. The Journal of Physical Chemistry, 88(20), 4451-4459.

Page 203: ﻡﻳﺣﺭﻟﺍ ﻥﺎﻣﺣﺭﻟﺍ ﷲ ﻡﺳﺑ NEW INHIBITORS OF …

184

Yan, J., Zhou, M., Gilbert, J. D., Wolff, J. J., Somogyi, Á., Pedder, R. E., Quintyn, R. S., Morrison, L. J., Easterling, M. L., Paša-Tolić, L., and Wysocki, V. H. (2017). Surface-induced dissociation of protein complexes in a hybrid fourier transform ion cyclotron resonance mass spectrometer. Analytical Chemistry, 89(1), 895-901.

Youngquist, R. S., Fuentes, G. R., Lacey, M. P., and Keough, T. (1995). Generation and screening of combinatorial peptide libraries designed for rapid sequencing by mass spectrometry. Journal of the American Chemical Society, 117(14), 3900-3906.

Zhang, H., Gu, Q., Liang, X., and Pan, Y. (2004). Screening for topoisomerase I binding compounds by high-performance liquid chromatography–mass spectrometry. Analytical Biochemistry, 329(2), 173-179.

Zhao, Y.-Z., van Breemen, R. B., Nikolic, D., Huang, C.-R., Woodbury, C. P., Schilling, A., and Venton, D. L. (1997). Screening solution-phase combinatorial libraries using pulsed ultrafiltration/electrospray mass spectrometry. Journal of Medicinal Chemistry, 40(25), 4006-4012.


Recommended