+ All Categories
Home > Documents > Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action,...

Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action,...

Date post: 02-Feb-2017
Category:
Upload: satya
View: 214 times
Download: 0 times
Share this document with a friend
12
1. Introduction 2. The gut microbiome and BSH-active bacterial cells 3. Probiotic bacteria as cholesterol-lowering therapeutics: in vitro, pre-clinical, and clinical evidence 4. Cholesterol-lowering mechanisms of action of BSH 5. Conclusion 6. Expert opinion Review Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications Mitchell L Jones, Catherine Tomaro-Duchesneau, Christopher J Martoni & Satya Prakash McGill University, Faculty of Medicine, Departments of Biomedical Engineering, Physiology, and Artificial Cells and Organs Research Center, Biomedical Technology and Cell Therapy Research Laboratory, Montreal, Quebec, Canada Introduction: Cardiovascular diseases (CVD) are the leading cause of global mortality and morbidity. Current CVD treatment methods include dietary intervention, statins, fibrates, niacin, cholesterol absorption inhibitors, and bile acid sequestrants. These formulations have limitations and, thus, additional treatment modalities are needed. Probiotic bacteria, especially bile salt hydrolase (BSH)-active probiotic bacteria, have demonstrated cholesterol-lowering efficacy in randomized controlled trials. Areas covered: This review describes the current treatments for CVD and the need for additional therapeutics. Gut microbiota etiology of CVD, cholesterol metabolism, and the role of probiotic formulations as therapeutics for the treatment and prevention of CVD are described. Specifically, we review studies using BSH-active bacteria as cholesterol-lowering agents with empha- sis on their cholesterol-lowering mechanisms of action. Potential limitations and future directions are also highlighted. Expert opinion: Numerous clinical studies have concluded that BSH-active probiotic bacteria, or products containing them, are efficient in lowering total and low-density lipoprotein cholesterol. However, the mechanisms of action of BSH-active probiotic bacteria need to be further supported. There is also the need for a meta-analysis to provide better information regarding the therapeutic use of BSH-active probiotic bacteria. The future of BSH-active probiotic bacteria most likely lies as a combination therapy with already existing treatment options. Keywords: bile salt hydrolase, cardiovascular disease, gut microbiota, heart health, hypercholesterolemia, hyperlipidemia, mechanism of action, microbiome, probiotic Expert Opin. Biol. Ther. (2013) 13(5):631-642 1. Introduction 1.1 Overview of hyperlipidemia and cardiovascular disease Cardiovascular disease (CVD), responsible for an estimated 16.7 million deaths worldwide, is the leading cause of global mortality and morbidity [1]. In Canada, 32.1% of all deaths in 2004 were due to CVD [2]. In 2007, 1.3 million Canadians were diagnosed with CVD by a health professional [2]. Among Canadians 75 years and older, 20.1% of women and 26.9% of men reported having 10.1517/14712598.2013.758706 © 2013 Informa UK, Ltd. ISSN 1471-2598, e-ISSN 1744-7682 631 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Biol. Ther. Downloaded from informahealthcare.com by UMEA University Library on 08/17/14 For personal use only.
Transcript
Page 1: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

1. Introduction

2. The gut microbiome and

BSH-active bacterial cells

3. Probiotic bacteria as

cholesterol-lowering

therapeutics: in vitro,

pre-clinical, and

clinical evidence

4. Cholesterol-lowering

mechanisms of action of BSH

5. Conclusion

6. Expert opinion

Review

Cholesterol lowering with bilesalt hydrolase-active probioticbacteria, mechanism of action,clinical evidence, and futuredirection for heart healthapplicationsMitchell L Jones, Catherine Tomaro-Duchesneau, Christopher J Martoni &Satya Prakash††McGill University, Faculty of Medicine, Departments of Biomedical Engineering, Physiology, and

Artificial Cells and Organs Research Center, Biomedical Technology and Cell Therapy Research

Laboratory, Montreal, Quebec, Canada

Introduction: Cardiovascular diseases (CVD) are the leading cause of global

mortality and morbidity. Current CVD treatment methods include dietary

intervention, statins, fibrates, niacin, cholesterol absorption inhibitors, and

bile acid sequestrants. These formulations have limitations and, thus,

additional treatment modalities are needed. Probiotic bacteria, especially

bile salt hydrolase (BSH)-active probiotic bacteria, have demonstrated

cholesterol-lowering efficacy in randomized controlled trials.

Areas covered: This review describes the current treatments for CVD and the

need for additional therapeutics. Gut microbiota etiology of CVD, cholesterol

metabolism, and the role of probiotic formulations as therapeutics for the

treatment and prevention of CVD are described. Specifically, we review

studies using BSH-active bacteria as cholesterol-lowering agents with empha-

sis on their cholesterol-lowering mechanisms of action. Potential limitations

and future directions are also highlighted.

Expert opinion: Numerous clinical studies have concluded that BSH-active

probiotic bacteria, or products containing them, are efficient in lowering

total and low-density lipoprotein cholesterol. However, the mechanisms of

action of BSH-active probiotic bacteria need to be further supported. There

is also the need for a meta-analysis to provide better information regarding

the therapeutic use of BSH-active probiotic bacteria. The future of

BSH-active probiotic bacteria most likely lies as a combination therapy with

already existing treatment options.

Keywords: bile salt hydrolase, cardiovascular disease, gut microbiota, heart health,

hypercholesterolemia, hyperlipidemia, mechanism of action, microbiome, probiotic

Expert Opin. Biol. Ther. (2013) 13(5):631-642

1. Introduction

1.1 Overview of hyperlipidemia and cardiovascular diseaseCardiovascular disease (CVD), responsible for an estimated 16.7 milliondeaths worldwide, is the leading cause of global mortality and morbidity [1].In Canada, 32.1% of all deaths in 2004 were due to CVD [2]. In 2007, 1.3 millionCanadians were diagnosed with CVD by a health professional [2]. AmongCanadians 75 years and older, 20.1% of women and 26.9% of men reported having

10.1517/14712598.2013.758706 © 2013 Informa UK, Ltd. ISSN 1471-2598, e-ISSN 1744-7682 631All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 2: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

CVD [2]. Economically, the direct and indirect costs of CVDhave reached $22.2 billion in Canada and $403.1 billion inthe United States [2]. A total of 162 million prescriptions werefilled in Canada for the treatment of CVD [2]. Coronaryartery disease (CAD), the most common form of CVD,is now the leading cause of death globally, as reported bythe World Health Organization (WHO), accounting for7.25 million deaths a year [3]. According to the present trends,one in two healthy males and one in three healthy females willdevelop CAD in their lifetime in the United States [4].Among confirmed independent risk factors, clinical

and epidemiological evidence have established a clear linkbetween elevated serum cholesterol and CAD [5-7]. Serumtotal cholesterol (TC) levels are correlated with CAD riskover a broad range of cholesterol values and in many popu-lations of the world [7]. Furthermore, evidence demonstratesa log-linear relationship between increasing low-densitylipoprotein cholesterol (LDL-C) concentration and arelative risk for CAD [8]. In addition, clinical trials forcholesterol-lowering therapies have generally confirmedthis relationship, showing an almost identical pattern ofassociation [9].This review highlights the need for additional cholesterol-

lowering therapeutics. We introduce the use of probioticformulations for the treatment and prevention of CVD. Wefocus on bile salt hydrolase (BSH)-active bacteria of the gutmicrobiota, and their use as cholesterol-lowering agents. Thecurrent evidence from in vitro, pre-clinical and clinicaldata is presented. A discussion on the cholesterol-loweringmechanisms of action of BSH-active probiotic bacteria isundertaken. Potential limitations and future directions arethen highlighted.

1.2 Current treatment modalities and limitationsThe primary target for the management of lipid levels innational and international guidelines remains LDL-C [10-12].Lower target cholesterol levels have also been recommendedfor patients with CAD, patients with very high CAD riskequivalents, and asymptomatic primary prevention patientswith multiple risk factors [11,13]. Dietary intervention is thefirst line of treatment; however, the effects are insufficientfor most individuals. Previous studies have shown that com-plete elimination of dietary cholesterol and limiting fat con-tent to < 10% of the daily caloric intake, results in only a4% regression of atherosclerotic plaques after 5 years, whencombined with stress management and aerobic exercise [14].

Several pharmacological agents are currently available forthe treatment of elevated LDL-C. These include 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins),fibric acids (fibrates), nicotinic acids (niacin), cholesterolabsorption inhibitors, and bile acid sequestrants. Due to along history of safety and efficacy, statins are the corner-stone of lipid-lowering therapy for reducing the levels ofLDL-C [15]. While statin therapy has been shown to signifi-cantly reduce the incidence of first or recurrent CAD-relatedevents, the majority of patients, especially those in the highestcategory of risk (individuals with the lowest LDL-C targets),fail to reach their goal on statin monotherapy [16-19]. Thesefindings are supported by the Lipid Treatment AssessmentProject (L-TAP), which found that 38% of patients treatedfor dyslipidemia, for a minimum of 3 months, attained theirNational Cholesterol Education Program (NCEP) LDL-Cgoals of which, only 18% with CAD reached these goals [19].Moreover, because of intolerance and safety concerns, patientsfrequently do not begin on or are titrated to an appropriatestatin dose [17,20]. Furthermore, the efficacy of statins islimited if stringent goals for serum LDL-C levels are notachieved in patients receiving statins alone or if side-effectsdevelop that require dose reduction or discontinuation [10,19].Studies have demonstrated that a doubling of the statin doseresulted in only a 6 -- 8% LDL-C reduction using currentlyavailable treatments [21,22]. Statins have also been found tobe less effective in lowering other lipid markers, such astriglycerides and lipoprotein(a), that are associated with therisk of atherosclerotic vascular disease [23,24]. Further, non-compliance to cholesterol-lowering medications was reportedin several clinical trials for primary and secondary prevention,with the risk of discontinuation shown to increase continu-ously over the treatment period, reaching 30% after 5 years [25].Thus, a wide gap between target and practice currently exists,and additional treatment modalities for LDL-C shouldbe explored.

1.3 Need for additional cholesterol-lowering

therapeuticsIn order to achieve a greater LDL-C reduction and, in turn,increase the percentage of patients attaining their NCEPLDL-C goal, it may be necessary to combine statins with

Article highlights.

. CVD, responsible for an estimated 16.7 million deathsworldwide, is the leading cause of global mortalityand morbidity.

. There is a growing interest in the use of probiotics,“live microorganisms which when administered inadequate amounts confer a health benefit on thehost,” for the treatment of CVD, specifically relatedto hypercholesterolemia.

. BSH activity of selected probiotic bacteria is associatedwith probiotic cholesterol-lowering properties inpreclinical and clinical trials, leading to decreased TC,LDL-C, non-HDL-C and apoB-100, with no significantside effects.

. There are a number of potential mechanisms of actionresponsible for BSH-active probiotic cholesterol lowering,including influencing bile acid binding of FXR anddecreased inward cholesterol transport throughABCG5/G8 and/or NPC1L1.

. BSH-active probiotic bacteria show potential, aloneor in combination with other cholesterol-loweringtherapeutics, for the treatment and prevention of CVD.

This box summarizes key points contained in the article.

M. L. Jones et al.

632 Expert Opin. Biol. Ther. (2013) 13(5)

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 3: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

agents that affect cholesterol through a different mechanismof action. For example, additive effects in reducing LDL-Cmay be achieved by combining a statin with either a bileacid sequestrant or a cholesterol absorption inhibitor (e.g.,ezetimibe), or with either of these classes plus niacin. Inpatients with elevated triglycerides, in those whom the pri-mary concern is pancreatitis rather than CAD, the combina-tion of a fibrate plus niacin or one of these agents plus fishoil (omega-3 fatty acids), or all three may be optimal [26].The combination of a statin with niacin or fenofibrate canprovide increased benefits in raising high-density lipoproteincholesterol (HDL-C) and reducing triglycerides in patientswith mixed dyslipidemias [26]. Niacin may also be added toan existing regimen to lower lipoprotein(a) levels. In additionto the potential for augmenting beneficial changes in lipidlevels, combination therapy may be based on the consider-ation that the use of lower doses of multiple drugs may resultin fewer or less severe side effects than the use of higher ormaximum doses of a single agent [26]. However, reports haveindicated that new drugs such as ezetimibe and torcetrapib,either have not shown an incremental reduction of secondaryendpoints in atherosclerotic disease, such as carotid intima-media thickness [27,28], or have had unanticipated adverseeffects [29]. Consequently, additional agents, such as pro-biotics, that target the metabolism of lipoproteins to improveoutcomes in patients with CVD could prove to be beneficial,with no or little risk for side effects.

1.4 Bile and bile saltsBile salts are synthesized from cholesterol in the liver, throughthe rate-limiting enzyme cholesterol 7a-hydroxylase(CYP7A1), and are the primary route of cholesterol excretionin the body. After synthesis, more than 99% of primary bileacids are conjugated either with the amino acid glycine ortaurine. The majority of bile salts are stored in the gallbladderand associated into mixed micelles containing bile salts,cholesterol and phosphatidylcholine. Upon gallbladder con-traction, often as a result of a meal, bile is released into theduodenum thus entering the enterohepatic circulation. Bilesalts have a number of functional roles, principally facilitatingthe intestinal digestion and absorption of dietary fats and fatsoluble vitamins [30]. Conjugated bile salts undergo severalimportant bacterial transformations in the gastrointestinaltract, including deconjugation and dehydroxylation by gutmicrobes, resulting in secondary bile salts such as deoxycho-late and lithocholate [31,32]. Bile salts are efficiently conservedin the enterohepatic circulation. After secretion into theintestine, the vast majority of bile salts are re-absorbed, eitherthrough active transport in the terminal ileum and possiblythe jejunum for conjugated bile salts, or passive diffusionin the small and large intestine for unconjugated bile salts [33].Following re-absorption, bile salts are transported through theportal circulation and returned to the liver, completing theenterohepatic circulation. In every bile cycle, approximately4% of bile salts are lost in the feces and de novo bile salt

synthesis from cholesterol is used to maintain the poolsize under steady state conditions. Bile salts are essential inmaintaining cholesterol homeostasis. Approximately 10% ofcholesterol is excreted in an unmetabolized form and evenless is utilized for other pathways, such as the production ofsteroid hormones and vitamin D [34]. Therefore, the conver-sion of cholesterol to bile salts in the liver and their subse-quent secretion and fecal excretion provides the major route(~ 90%) for the elimination of excess cholesterol. Hence,one can speculate that probiotic bacteria, that target themetabolism of bile salts, could prove beneficial to patientswith CVD.

1.5 Probiotic bacteriaOver the past two decades, there has been a growing interestin both the basic and clinical science of probiotics, whichare defined by the WHO as “live microorganisms whichwhen administered in adequate amounts confer a healthbenefit on the host,” and are being examined for their efficacyin preventing or treating a host of diseases [35,36]. This interesthas resulted in more than 6000 publications in the biomedicalliterature, with over 60% published since 2008 [37]. The inter-est in probiotics and GI microbiota has been echoed in themedia [38] and the business [39] worlds. The human GI tracthouses over 1014 bacterial cells, the most numerically pre-dominant of which are from the Firmicutes and Bacteroidetesphyla, with the most abundant bacteria from the Bacteroidesgenus [35,40,41]. Probiotic strains belong mostly to the generaLactobacillus and Bifidobacterium. Evidence from over700 randomized, placebo-controlled, clinical trials has shownthat certain probiotic bacterial strains may aid in: preventingmetabolic syndrome [40,42], reducing the duration of acutediarrhea [43,44], reducing the risk of nosocomial diarrhea androtavirus gastroenteritis [45], preventing sepsis and severe acutepancreatitis [46], reducing post-operative infection rate [46,47],preventing periodontal diseases [48], preventing allergy andreducing colonization by bacterial pathogens [49], improvingsymptomatology of inflammatory bowel disease [50] andreducing lipids in hyperlipidemic adults [51-56].

2. The gut microbiome and BSH-activebacterial cells

The gut microbiome plays a crucial role in human health anddisease [35,57]. Interestingly, the gut microbiome, whichincludes all gut microorganisms, their genetic informationand their environmental interactions, is 150 times largerthan the human genome [58]. The gut microbiota’s relation-ship with the epithelial layers of the GI and the gut-associatedlymphoid tissue highlights the symbiotic relationship betweenthe microbiota and the eukaryotic host [59]. The gut micro-biome has evolved to perform metabolic functions otherwisenot performed by human eukaryotic cells. For example,early research demonstrated that the synthesis of riboflavin,vitamin B, pantothenic acid, vitamin B12, folic acid,

Cholesterol lowering by bile salt hydrolase-active probiotic bacteria: mechanism, clinical evidence and heart health applications

Expert Opin. Biol. Ther. (2013) 13(5) 633

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 4: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

nicotinic acid, thiamine and biotin is performed by the GImicrobiota [60].It was first observed in the 1960s that germ-free animals

accumulated cholesterol in greater quantities and at a fasterrate than their conventionally raised counterparts [61]. The pro-posed explanation was that germ-free animals catabolize choles-terol at a slower rate, and that the intestinal microbiota wasresponsible for accelerating cholesterol catabolism through anincrease in the elimination of bile acids [62]. This was furthersubstantiated when it was shown that germ-free animals haveelevated levels of conjugated bile acids throughout the intestine,significantly reduced fecal biliary excretion and three times thebile acid concentration in bile [63-67].More recently, comprehensive porcine studies showed that,

by increasing the total BSH activity of the GI microbiota, asignificant increase in the deconjugated bile acid pool wasobserved [68,69]. BSH activity is responsible for the deconjuga-tion of bile acids, by hydrolysis of the amide bond, of the con-jugated bile acid, and liberation of the glycine/taurine moietyfrom the steroid core. In addition, the GI microbiota hasbeen shown as responsible for the elimination of sterols, withthe intestinal flora promoting cholesterol catabolism in conven-tional, as compared to germ-free animals [62]. BSH activity isspecific to the microbiota and is not present in eukaryotic cells,substantiating the importance of the gut microbiota incholesterol metabolism. BSH activity has been characterizedin Lactobacillus [70-73], Bifidobacterium [74-77], Clostridium [78],Enterococcus [79] and Bacteroides [80,81]. Functions of BSH inbacterial cells are still not completely understood, but thereare several hypotheses. Potential roles, discussed in a previousreview, include nutrition, cell surface modifications, biledetoxification and GI persistence [82]. In terms of cholesterolmetabolism, mice treated with a three-day course of oral anti-biotics increased biliary bile acid output threefold while fecaloutput decreased by 70% [83]. These findings support thehypothesis that the intestinal microbiota specifically increaseddeconjugation of intestinal bile acids by BSH, and increasedlevels of deconjugated bile acids entering the enterohepaticcirculation and via the portal vein.

3. Probiotic bacteria as cholesterol-loweringtherapeutics: in vitro, pre-clinical, andclinical evidence

3.1 Cholesterol-lowering with probioticsProbiotic bacteria have been investigated for their clinicalefficacy in lowering LDL-C. A crossover study, in 30 normoli-pidemic male participants, reported a decrease in TC andLDL-C by 4.4% and 5.4%, respectively, following the con-sumption of yogurt enriched with Lactobacillus acidophilusand fructooligosaccharides three times daily [55]. A rando-mized, blinded, clinical study using 32 participants withmild to moderate hypercholesterolemia, reported a decreasein TC and LDL-C by 5.3% and 6.15%, respectively, follo-wing consumption of a fermented milk product containing

Enterococcus faecium [53]. In addition, a randomized, blinded,study in 58 non-obese, normocholesterolemic middle-agedmen, reported a decrease in LDL-C by 10% following theconsumption of a fermented milk product containingE. faecium and two strains of Streptococcus thermophilus [51].On the other hand, several placebo-controlled studies havereported little or no effect following the daily consumptionof various supplements and foods containing probioticbacteria [84-90]. These contradictory results can be largelyattributed to inadequate strain selection, small sample size orpoor clinical design [91]. In an attempt to increase statisticalpower, a recent meta-analysis integrating results from 13 indi-vidual lipid-lowering probiotic clinical studies [92] demon-strated a LDL-C mean net change of -0.12 mmol/L (95%confidence interval: -0.206 to -0.049, p < 0.01) or approxi-mately 3%. A number of research groups have also shownthat BSH-active probiotic bacteria can be used to lowercholesterol in murine models [93-95].

3.2 Cholesterol lowering with BSH-active probioticsRecently, a randomized, placebo-controlled clinical trialshowed a significant reduction in plasma TC and LDL-C asa result of the administration of BSH-active L. acidophilusCHO-220 combined in a capsule with the prebiotic inulin [54].The BSH phenotype of selected probiotic strains can be asso-ciated with the capacity of an organism to reduce cholesterollevels, as demonstrated in pre-clinical and clinical studies, aswell as improved survival of the organism in the GI tract, asBSH is associated with bile tolerance [96]. In addition, ourgroup has previously published an extensive in vitro characte-rization of Lactobacillus reuteri NCIMB 30242 using a combi-nation of molecular and metabolic analysis techniques tosupport its safe use as a lipid-lowering probiotic inhumans [97]. We have performed a double-blind, randomized,placebo-controlled, parallel-arm, multi-center study to evalu-ate the cholesterol-lowering effect of microencapsulatedBSH-active L. reuteri NCIMB 30242 (~ 1 � 1010 CFU/yogurt), in yogurt format, over 6 weeks [56]. Subjects con-suming the probiotic yogurts attained significant reductionsin LDL-C of 8.92%, TC of 4.81% and non-HDL-C of6.01%, as compared to placebo [56]. There was also a signifi-cant absolute change in apolipoprotein B-100 (apoB-100)of -0.19 mmol/L over the 6-week treatment period [56].Importantly, the placebo and treatment groups remainedcomparable for biomarkers of safety at the study endpoint [56].In a second double-blind, randomized, placebo-controlled,parallel-arm, multi-center study, we have evaluated the safetyand efficacy of an improved L. reuteri NCIMB 30242 capsuleformulation (~ 3 � 109 CFU/capsule) over 9 weeks [98].Subjects consuming the probiotic capsules attained significantreductions in LDL-C of 11.64%, TC of 9.14%, non-HDL-Cof 11.30% and apoB-100 of 8.41%, over the 9-week treat-ment period, as compared to placebo. Additionally, signifi-cant reductions in cardiovascular risk factors were observedfor fibrinogen of 14.24% and high-sensitivity-C-reactive

M. L. Jones et al.

634 Expert Opin. Biol. Ther. (2013) 13(5)

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 5: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

protein (hs-CRP) of 192.92%. In support of the proposedmechanism of action, we found significant increases in decon-jugated bile acids, indicating increased intraluminal BSHactivity, and significant reductions in the surrogate absorptionof sterols, indicating a reduction in inward cholesteroltransport [98]. A comparison of individual LDL-C responses,at the study endpoint, indicates that, despite a proportion ofsubjects that experienced unchanged LDL-C values, a generalLDL-C lowering effect was observed. In addition, weobserved significant reductions in cholesterol esters, whichindicated a reduction in the storage form of cholesterol seenin foamy cells and atherosclerotic plaques. Finally, no adverseevents or biochemical safety endpoints were determined to besubstantial or associated with the treatment [99,100].

4. Cholesterol-lowering mechanisms ofaction of BSH

There are a number of mechanisms of action potentiallyinvolved in lowering cholesterol by BSH-active probioticbacteria, as shown in Figure 1. BSH-active probiotic bacteria

may act in the lumen of the intestine as well as in the liverand other organs. BSH-active probiotics have been shownto increase intraluminal bile acid deconjugation, resultingin increased levels of circulating deconjugated bile salts inhumans [56] and pigs [69]. The increased deconjugation ofbile salts decreases cholesterol absorption by enterocytes,simply due to a decrease in primary and secondary bileacids and the micellization capacity of conjugated andunconjugated bile salts.

In addition, by modifying the bile acid pool profile,BSH-active bacteria may influence the farnesoid X receptor(FXR), a bile acid nuclear receptor [56,101]. Endogenous bileacids activate FXR with different efficacy (chenodeoxycholicacid > lithocholic acid = deoxycholic acid > cholic acid), withthe conjugated forms being less potent [102]. It has been shownthat decreased FXR activity results in the downregulation ofsmall heterodimer partner (SHP) and increased catabolism ofcholesterol and synthesis of bile acids by CYP7A1 [101]. Thedownregulation of SHP results in the upregulation of the liverX receptor (LXR) [101]. This, in turn, leads to the upregulationof adenosine triphosphate-binding cassette transporters G5 and

LDL-Receptors

LDL-C (Blood)

7α-hydroxylase

BA (Hepatocyte)

Bile acid(CBA + BA)

(Enterohepatic)

FXR SHP

CYP7A1

LXR ABCG5/G8

BA

Cholesterol(Hepatocyte)

LDL-Receptor

ABCA1ABCG1

ABCA1ABCG1

ABCG5/G8

BSH-activeLactobacillus

ApoA-1ABCA1ABCG1

NPC1L1

LXR

BA and cholesterolexcretion (Feces)

CBA BA

BSH

(GI lumen)

SR-BIApoA-1

HDL-CCholesterol

(Macrophage)Cholesterol(Enterocyte)

CholesterolBile

(Biliary)

Cholesterol(GI lumen)

LDL-C(Blood)

Cholesterol(Hepatocyte)

BA

H

C

O

HN H

HDCA

H2O+

OH

HO

DCA

H

C OH + OH

OH

HO

Hydrolysis

Bile salt hydrolase (BSH)

O

H2N

S O

O

O

OHH2N

O

Figure 1. Schematic representation of the potential effects of probiotic BSH activity on cholesterol metabolic pathways. BSH

enzymatic activity hydrolyzes conjugated bile acids (CBA) to deconjugated bile acids (BA).

Cholesterol lowering by bile salt hydrolase-active probiotic bacteria: mechanism, clinical evidence and heart health applications

Expert Opin. Biol. Ther. (2013) 13(5) 635

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 6: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

G8 (ABCG5/G8) [103]. The upregulation of ABCG5/G8results in a decrease in the intestinal absorption of cholesterol,while promoting their biliary excretion. Support for thishypothesis can be found in the finding that the oral feedingof Lactobacillus plantarum KCTC3928 to mice resulted in sig-nificant LDL-C and triglyceride lowering effects, increasedfecal bile acid excretion, increased hepatic bile acid synthesisand increased expression of CYP7A1, enhancing cholesterolcatabolism and bile synthesis [93]. Also, it was found that theoverexpression of ABCG5/G8 in transgenic mice limits sterolabsorption and promotes neutral sterol excretion [104]. Further-more, the deconjugated bile acids preferentially increaseATPase activities of ABCG5/G8 transporters found on theapical membranes of enterocytes and hepatocytes, limitingthe accumulation of cholesterol by transporting it into theintestinal lumen and bile [105]. In addition, BSH-active Lactoba-cillus, using Caco-2 cells, resulted in the upregulation of LXRconcomitantly with the elevated expression of ABCG5/G8heterodimeric transporters, and the promotion of cholesterol

efflux, with no noticeable effects on Niemann-Pick C1 Like 1(NPC1L1) expression [103].

Another proposed mechanism of action associated withBSH activity involves the inhibition of NPC1L1, similarto plant sterols [106]. It has also been demonstrated thatBSH activity and increased circulating deconjugated bileresult in significant reductions in serum plant sterols,surrogate markers for cholesterol absorption. This is similarto the mechanism of action of the drug ezetimibe, a potentcholesterol absorption inhibitor [107]. NPC1L1 is responsiblefor the bulk movement of cholesterol into the enterocytes,with genetic inactivation resulting in a significant reductionof cholesterol absorption [108]. Indeed, a decrease inNPC1L1 was demonstrated in rats administered a BSH-active L. acidophilus [109,110]. In addition, in recent research,Yoon et al. have demonstrated a downregulation ofNPC1L1 in Caco-2 colon epithelial cells by Lactobacillusrhamnosus and L. plantarum, as well as increased cholesterolefflux in macrophages [111,112].

Table 1. Potential effects of BSH-active bacteria on cholesterol metabolic pathways.

BSH-active probiotic targets BSH-active probiotic

effect on target

Impact of modified expression on

cholesterol metabolic pathways

Refs.

ATP binding cassette A1 (ABCA1) Upregulated Increased cholesterol efflux to lipoproteinsIncreased plasma HDL-C levels

[110,112]

ATP binding cassette G1 (ABCG1) Upregulated Increased cholesterol efflux [110,112]

ATP binding cassette G5/G8 (ABCG5/G8) Upregulated Increased biliary excretionIncreased cholesterol efflux

[103,105]

Cholesterol 7 alpha-hydroxylase (CYP7A1) Upregulated Increased cholesterol catabolismIncreased bile acid synthesisReduced cholesterol levels in hepatocytes

[93]

Farnesoid X receptor (FXR) Downregulated Decreased activation of SHPIncreased activation of CYP7A1

[93]

Liver X receptor (LXR) Upregulated Increased activation of CYP7A1Increased ABCG5/G8Increased activation of ABCA1Increased activation of ABCG1

[106,112]

Niemann-Pick C1-like 1 (NPC1L1) Downregulated Upregulation of HMG-CoA synthaseDecreased intestinal cholesterol absorptionUpregulation of ABCA1

[106,109-111]

Small heterodimer partner (SHP) Downregulated Increased activation of CYP7A1 [101]

Table 2. Effective dose and cholesterol reduction by BSH-active Lactobacillus and other nutritional ingredients.

Effective

dose (g/day)*

TC reduction vs

placebo (%)

LDL-C reduction vs

placebo (%)

LDL-C reduction vs

placebo (Median %/g

effective dose)

Refs.

BSH-active Lactobacillus 0.2 5 -- 9 9 -- 12 52.5 [54,56,98]§

Phytosterolsz 2 -- 3 3 -- 15 5 -- 15 4.0 [121-125]

Beta-glucanz 3 4 -- 9 5 -- 10 2.5 [126-129]

Psylliumz 7 2 -- 9 3 -- 7 1.4 [130,131]

Soy proteinz 25 3 -- 5 4 -- 6 0.2 [132-135]

*Effective dose in grams of dry ingredient.zApproved Food and Drug Administration (FDA) heart health claim.§Study performed with BSH-active Lactobacillus and prebiotic inulin [54].

M. L. Jones et al.

636 Expert Opin. Biol. Ther. (2013) 13(5)

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 7: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

In addition, a recent study by Jones et al. demonstrated thatincreased circulating bile was correlated with a reductionin serum LDL-C and apoB-100 [56]. ApoB-100 is found innon-HDL-C cholesterol originating from the liver, and itslevels are well-correlated with CAD [113]. ApoB-100 is aprimary structural protein of the atherogenic lipoproteins,and so its decrease results in reducing LDL-C serum levels.Indeed, Sniderman et al. have demonstrated an inverse rela-tion between bile acid synthesis and apoB-100 secretion [114].This could point to an alternate mechanism of actionof bacterial BSH activity. Of interest is the fact that thecholesterol-reducing drug-candidate Mipomersen, an anti-sense oligonucleotide, acts on a similar mechanism of actioninvolving the inhibition of apoB-100 synthesis in the liverthrough RNA interference technology [115].

Despite a growing understanding of the mechanismsthrough which increased deconjugation of bile may lead toincreased cholesterol catabolism, reduced cholesterol absorp-tion and reduced serum cholesterol, detailed studies arerequired to elucidate the precise mechanisms of action.

5. Conclusion

CVD is the leading cause of global mortality and morbidity,but current treatment modalities are inefficient and presentimportant side effects and toxicity issues. Using germ-free animal studies, the role of the GI microbiota in choles-terol metabolism was demonstrated. A number of probioticbacteria have been successfully used as cholesterol-loweringbiotherapeutics. BSH, an enzyme expressed solely by bacteria,including a number of probiotic bacteria, is responsible forthe deconjugation of bile salts in the intestinal lumen, limitingthe absorption of cholesterol through the enterocytes. Theadministration of BSH-active probiotic bacteria has demon-strated significant cholesterol-lowering effects in vitro and inpreclinical and human clinical studies. There are a numberof hypothesized mechanisms of action by which the BSH-active probiotic bacteria may be exerting their cholesterol-lowering effects. Players of the cholesterol metabolic pathwaysthat may be modulated by probiotic BSH activity include, butare not limited to, FXR, SHP, CYP7A1, LXR, ABCG5/G8,NPC1L1 and apoB-100. Despite the abundance of existingdata on the beneficial effects of BSH-active probioticbacteria and their modulation of important regulatorsinvolved in cholesterol metabolism, detailed mechanisticstudies are nonetheless required for a complete understandingof the cholesterol-lowering effects. This is crucial for thefurther optimization and development of a probioticcholesterol-lowering therapeutic.

6. Expert opinion

Probiotic bacteria have gained much interest in recent yearsfor the prevention and treatment of a number of healthdisorders. BSH-active probiotic bacteria have shown promise

for their cholesterol-lowering properties and as a potentialadjunct therapy for the management of CVD, in several largerandomized placebo-controlled clinical trials. As described inthis review, an important consideration for BSH-active probi-otics is the need for a better understanding of the mechanismsof action responsible for the observed cholesterol-loweringeffects. Table 1 presents a list of hypothesized targets ofBSH-active probiotic bacteria for their cholesterol-loweringeffects. A second consideration is safety, as with the deliveryof all live probiotic microorganisms. On this point, clinicaltoxicological studies have been published confirming thesafety and tolerability of BSH-active probiotic bacteria [99,100].These studies have reported no significant changes inhematological markers of safety, biochemical markers ofsafety or adverse effects [56]. Moreover, an improvement ingastrointestinal symptoms, including GI discomfort, bloating,constipation and diarrhea, was detected by a Rome III GIquestionnaire following BSH-active probiotic administra-tion [116]. Of interest is the comparison of BSH-active probi-otics with available nutritional supplements for cholesterollowering, specifically in terms of a significantly lower effectivedose, as presented in Table 2.

The potential link between widespread antibiotic use and thehigh prevalence of hypercholesterolemia in Western societies isalso of interest. The intestinal microbiota plays an importantrole in the metabolism of cholesterol by interacting with bileacids and cholesterol. In the feces of man and other mammals,the major microbial cholesterol derivative is coprostanol. Itis well-established that germ-free animals lack cholesterol-metabolizing microbes and do not have an intestinal microbialexcretion pathway for cholesterol. As a result, they have reducedde novo synthesis of cholesterol and higher serum cholesterollevels than their conventional counterparts [117]. Antibiotic usehas been shown to permanently change the GI microbiotaresulting in significantly reduced diversity, as well as reducedconversion of cholesterol to coprostanol. Hence, althoughthe Western diet is commonly cited as a causative factor forhypercholesterolemia, reduced BSH activity due to widespreadantibiotic use may also play a causative role and should be inves-tigated. A metagenomic analysis of patients exposed to anti-biotics compared to naive patients should be examined toevaluate the relative quantity and conservation of the BSHphenotype following antibiotic use.

BSH-active probiotic formulations may have potential forcholesterol lowering in combination with plant sterols,omega-3 fatty acids, fish oils, and statins. Plant sterols act bycompetitively inhibiting cholesterol absorption by NPC1L1transporters and have been shown to increase levels of serumplant sterols [118]. Recently, plant sterols have been foundas part of atherosclerotic plaques [119] and in the retina oflong-term plant sterol and stanol users [120]. BSH-activeprobiotic bacteria are postulated to function by inhibitingthe absorption and/or increasing the excretion of neutralsterols, likely through the upregulation and increased activityof ABCG5/G8 transporters. In addition, they have been

Cholesterol lowering by bile salt hydrolase-active probiotic bacteria: mechanism, clinical evidence and heart health applications

Expert Opin. Biol. Ther. (2013) 13(5) 637

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 8: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

shown to reduce circulating cholesterol and plant sterols [98].Thus, a combination of plant sterols and BSH-active pro-biotics should be evaluated for an improved cholesterol-lowering effect while reducing plant sterol serum levels,as well as potentially improving the safety profile of sterolproducts. On the other hand, fish oils, omega-3 fatty acids,medium chain triglycerides, and alpha-linoleic acids aimat improving the ratio of ‘good’ to ‘bad’ fatty acids in theblood. The result of consuming beneficial fatty acids is thatthe esterified cholesterol profile is improved by increasingthe proportion of monounsaturated and polyunsaturatedcholesterol esters at the expense of saturated fatty acidcholesterol esters, a shift that has been correlated with a lowerrisk of atherosclerosis and cardiovascular events. A com-bination of beneficial fatty acids and BSH-active probioticsmay result in a significant decrease in serum cholesterol whileimproving the relative molar fraction of beneficial fattyacids esterified to cholesterol. Finally, it is postulated thatBSH-active bacteria would work in a complementary fashionwith statins to amplify LDL receptor activity and the clear-ance of serum cholesterol [56], as they increase bile salt decon-jugation and reduce sterol absorption. Therefore, a greaterreduction in serum LDL-C is hypothesized when admini-stering BSH-active probiotic bacteria concomitantly to statin

monotherapy. Well-designed randomized controlled clinicalstudies should be performed to investigate the use of suchcombination therapies. There is also a need for meta-analyses to better examine the variation in study outcomesbetween studies. Factors that may impact on outcomes ofcholesterol-lowering strategies with BSH-active probioticbacteria must also be examined.

Acknowledgments

The authors would like to acknowledge a Canadian Instituteof Health Research (CIHR) Grant (MPO 64308) andgrants from Micropharma Ltd. to S Prakash and a DoctoralAlexander Graham Bell Canada Graduate Scholarship fromthe Natural Sciences and Engineering Research Councilof Canada (NSERC) to C Tomaro-Duchesneau. ML Jonesand C Tomaro-Duchesneau contributed equally to this work.

Declaration of interest

ML Jones and S Prakash are co-founders and shareholders ofMicropharma Ltd and report a conflict of interest. CJ Martoniis employed by and is a shareholder of Micropharma Ltd.C Tomaro-Duchesneau reports no conflict of interest.

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Tarride JE, Lim M, DesMeules M, et al.

A review of the cost of cardiovascular

disease. Can J Cardiol 2009;25:e195-202

2. Public Health Agency of Canada.

Tracking Heart Disease and Stroke in

Canada, 2009

3. World Health Organization. The top

10 causes of death. Fact Sheet No 310,

2011

4. Durrington P. Dyslipidaemia. Lancet

2003;362:717-31

5. The lipid research clinics coronary

primary prevention trial results. I.

reduction in incidence of coronary heart

disease. JAMA 1984;20:351-64

6. Muldoon MF, Manuck SB,

Matthews KA. Lowering cholesterol

concentrations and mortality:

a quantitative review of primary

prevention trials. BMJ 1990;301:309-14

7. Ridker PM. Evaluating novel

cardiovascular risk factors: can we better

predict heart attacks? Ann Intern Med

1999;130:933-7

8. Jacobson TA. “The lower the better” in

hypercholesterolemia therapy: a reliable

clinical guideline? Ann Intern Med

2000;133:549-54

9. Hunninghake DB. Therapeutic efficacy

of the lipid-lowering armamentarium: the

clinical benefits of aggressive

lipid-lowering therapy. Am J Med

1998;104:9S-13S

10. National Cholesterol Education Program

(NCEP) Expert Panel on Detection,

Evaluation, and Treatment of High

Blood Cholesterol in Adults (Adult

Treatment Panel III). Circulation

2002;106:3143-421

11. Grundy SM, Cleeman JI, Merz CNB,

et al. Implications of recent clinical trials

for the National Cholesterol Education

Program Adult Treatment Panel III

guidelines. Circulation 2004;110:227-39

12. Genest J, Frohlich J, Fodor G,

McPherson R. Recommendations for the

management of dyslipidemia and the

prevention of cardiovascular disease:

summary of the 2003 update. CMAJ

2003;169:921-4

13. Smith SC, Allen J, Blair SN, et al. AHA/

ACC guidelines for secondary prevention

for patients with coronary and other

atherosclerotic vascular disease:

2006 update. Circulation

2006;113:2363-72

14. Ornish D, Brown SE, Billings JH, et al.

Can lifestyle changes reverse coronary

heart disease? Lancet 1990;336:129-33

15. Brautbar A, Ballantyne CM.

Pharmacological strategies for lowering

LDL cholesterol: statins and beyond.

Nat Rev Cardiol 2011;8:253-65. Update on LDL-C-lowering agents

available or under development.

16. Andrews TC, Ballantyne CM, Hsia JA,

Kramer JH. Achieving and maintaining

national cholesterol education program

low-density lipoprotein cholesterol goals

with five statins. Am J Med

2001;111:185-91

17. Foley KA, Simpson J, Crouse JR III,

et al. Effectiveness of statin titration on

low-density lipoprotein cholesterol goal

attainment in patients at high risk of

atherogenic events. Am J Cardiol

2003;92:79-81

18. Ford ES, Mokdad AH, Giles WH,

Mensah GA. Serum total cholesterol

concentrations and awareness, treatment,

and control of hypercholesterolemia

among US adults. Circulation

2003;107:2185-9

19. Pearson TA, Laurora I, Chu H,

Kafonek S. The lipid treatment

assessment project (l-tap): a multicenter

M. L. Jones et al.

638 Expert Opin. Biol. Ther. (2013) 13(5)

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 9: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

survey to evaluate the percentages of

dyslipidemic patients receiving

lipid-lowering therapy and achieving

low-density lipoprotein cholesterol goals.

Arch Intern Med 2000;160:459-67

20. Frolkis JP, Zyzanski SJ, Schwartz JM,

Suhan PS. Physician noncompliance with

the 1993 National Cholesterol Education

Program (NCEP-ATPII) guidelines.

Circulation 1998;98:851-5

21. Davidson MH, Palmisano J, Wilson H,

et al. A multicenter, randomized,

double-blind clinical trial comparing the

low-density lipoprotein

cholesterol-lowering ability of lovastatin

10, 20, and 40 mg/d with fluvastatin

20 and 40 mg/d. Clin Ther

2003;25:2738-53

22. Jones P, Kafonek S, Laurora I,

Hunninghake D. Comparative dose

efficacy study of atorvastatin versus

simvastatin, pravastatin, lovastatin, and

fluvastatin in patients with

hypercholesterolemia (The CURVES

Study). Am J Cardiol 1998;81:582-7

23. Sarwar N, Danesh J, Eiriksdottir G,

et al. Triglycerides and the risk of

coronary heart disease. Circulation

2007;115:450-8

24. Bennet A, Di Angelantonio E, Erqou S,

et al. Lipoprotein(a) levels and risk of

future coronary heart disease: large-scale

prospective data. Arch Intern Med

2008;168:598-608

25. Insull W. The problem of compliance to

cholesterol altering therapy. J Intern Med

1997;241:317-25

26. Jones PH. Statins as the cornerstone of

drug therapy for dyslipidemia:

monotherapy and combination therapy

options. Am Heart J 2004;148:S9-S13

27. Kastelein JJP, Akdim F, Stroes ESG,

et al. Simvastatin with or without

ezetimibe in familial

hypercholesterolemia. N Engl J Med

2008;358:1431-43

28. Taylor AJ, Villines TC, Stanek EJ, et al.

Extended-release niacin or ezetimibe and

carotid intima-media thickness. N Engl

J Med 2009;361:2113-22

29. Barter PJ, Caulfield M, Eriksson M,

et al. Effects of torcetrapib in patients at

high risk for coronary events. N Engl

J Med 2007;357:2109-22

30. Hofmann AF. Bile acid secretion, bile

flow and biliary lipidttsecretion in

humans. Hepatology 1990;12:17S-22S

31. Midtvedt T. Microbial bile acid

transformation. Am J Clin Nutr

1974;27:1341-7

32. Jonsson G, Midtvedt AC, Norman A,

Midtvedt T. Intestinal microbial bile acid

transformation in healthy infants.

J Pediatr Gastroenterol Nutr

1995;20:394-402

33. Trauner M, Boyer JL. Bile salt

transporters: molecular characterization,

function, and regulation. Physiol Rev

2003;83:633-71

34. Hofmann AF. Bile acids: the good, the

bad, and the ugly. Physiology

1999;14:24-9

35. Prakash S, Tomaro-Duchesneau C,

Saha S, Cantor A. The gut microbiota

and human health with an emphasis on

the use of microencapsulated bacterial

cells. J Biomed Biotechnol 2011;2011:12. Comprehensive review on the use of

probiotics for a number of

health disorders.

36. Health and nutritional properties of

probiotics in food including powder milk

with live lactic acid bacteria. FAO,

WHO, Argentina; 2001

37. Rijkers GT, de Vos WM, Brummer R-J,

et al. Health benefits and health claims

of probiotics: bridging science and

marketing. Br J Nutr 2011;106:1291-6

38. Antibiotics in early infancy could raise

obesity risk: study. CTV News Canada.

2012. Available from: http://www.

ctvnews.ca/health/antibiotics-in-early-

infancy-could-raise-obesity-risk-study-

1.923213

39. Microbes maketh man. The Economist

2012; August 18th

40. Wallace TC, Guarner F, Madsen K,

et al. Human gut microbiota and its

relationship to health and disease.

Nutr Rev 2011;69:392-403

41. Arumugam M, Raes J, Pelletier E, et al.

Enterotypes of the human gut

microbiome. Nature 2011;473:174-80

42. Bhathena J, Tomaro-Duchesneau C,

Martoni C, et al. Effect of orally

administered microencapsulated

FA-producing L. fermentum on markers

of metabolic syndrome: an in vivo

analysis. J Diabetes Metab 2012;S2(009)

43. Huang JS, Bousvaros A, Lee JW, et al.

Efficacy of probiotic use in acute

diarrhea in children: a meta-analysis.

Dig Dis Sci 2002;47:2625-34

44. Szajewska H, Mrukowicz JZ. Probiotics

in the treatment and prevention of acute

infectious diarrhea in infants and

children: a systematic review of published

randomized, double-blind,

placebo-controlled trials. J Pediatr

Gastroenterol Nutr 2001;33:S17-25

45. Szajewska H, Kotowska M,

Mrukowicz JZ, et al. Efficacy of

lactobacillus GG in prevention of

nosocomial diarrhea in infants. J Pediatr

2001;138:361-5

46. Gill HS, Guarner F. Probiotics and

human health: a clinical perspective.

Postgrad Med J 2004;80:516-26

47. Rayes N, Seehofer D, Hansen S, et al.

Early enteral supply of lactobacillus and

fiber versus selective bowel

decontamination: a controlled trial in

liver transplant recipients.

Transplantation 2002;74:123-7

48. Saha S, Tomaro-Duchesneau C,

Tabrizian M, Prakash S. Probiotics as

oral health biotherapeutics. Expert Opin

Biol Ther 2012;12:1207-20

49. Lodinova-Zadnikova R, Sonnenborn U.

Effect of preventive administration of a

nonpathogenic Escherichia coli strain on

the colonization of the intestine with

microbial pathogens in newborn infants.

Neonatology 1997;71:224-32

50. Hedin CRH, Mullard M, Sharratt E,

et al. Probiotic and prebiotic use in

patients with inflammatory bowel disease:

a case-control study. Inflamm Bowel Dis

2010;16:2099-108

51. Agerbaek M, Gerdes LU, Richelsen B.

Hypocholesterolaemic effect of a new

fermented milk product in healthy

middle-aged men. Eur J Clin Nutr

1995;49:346-52

52. Agerholm-Larsen L, Raben A,

Haulrik N, et al. Effect of 8 week intake

of probiotic milk products on risk factors

for cardiovascular diseases. Eur J

Clin Nutr 2000;54:288-97

53. Bertolami MC, Faludi AA, Batlouni M.

Evaluation of the effects of a new

fermented milk product (Gaio) on

primary hypercholesterolemia. Eur J

Clin Nutr 1999;53:97-101

54. Ooi LG, Ahmad R, Yuen KH,

Liong MT. Lactobacillus acidophilus

CHO-220 and inulin reduced plasma

total cholesterol and low-density

lipoprotein cholesterol via alteration of

Cholesterol lowering by bile salt hydrolase-active probiotic bacteria: mechanism, clinical evidence and heart health applications

Expert Opin. Biol. Ther. (2013) 13(5) 639

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 10: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

lipid transporters. J Dairy Sci

2010;93:5048-58

55. Schaafsma G, Meuling WJ,

van Dokkum W, Bouley C. Effects

of a milk product, fermented by

Lactobacillus acidophilus and with

fructo-oligosaccharides added, on blood

lipids in male volunteers. Eur J

Clin Nutr 1998;52:436-40

56. Jones ML, Martoni CJ, Parent M,

Prakash S. Cholesterol-lowering efficacy

of a microencapsulated bile salt

hydrolase-active Lactobacillus reuteri

NCIMB 30243 yoghurt formulation in

hypercholesterolaemic adults. Br J Nutr

2012;107:1505-13.. Study investigating a BSH-active

probiotic for cholesterol lowering.

57. Cani PD. The role of the gut microbiota

in energy metabolism and metabolic

disease. Curr Pharm Des

2009;15:1546-58

58. Lepage P, Leclerc MC, Joossens M, et al.

A metagenomic insight into our gut’s

microbiome. Gut 2012: in press

59. Hord NG. Eukaryotic-microbiota

crosstalk: potential mechanisms for health

benefits of prebiotics and probiotics.

Annu Rev Nutr 2008;28:215-31

60. Coates ME, Ford JE, Harrison GF.

Intestinal synthesis of vitamins of the

B complex in chicks. Br J Nutr

1968;22:493-500

61. Kellogg TF, Wostmann BS. The

response of germfree rats to dietary

cholesterol. Adv Exp Med Biol

1969;3:293

62. Wostmann BS, Wiech NL, Kung E.

Catabolism and elimination of

cholesterol in germfree rats. J Lipid Res

1966;7:77-82

63. Madsen D, Beaver M, Chang L, et al.

Analysis of bile acids in conventional and

germfree rats. J Lipid Res

1976;17:107-11

64. Jones BV, Begley M, Hill C, et al.

Functional and comparative

metagenomic analysis of bile salt

hydrolase activity in the human gut

microbiome. Proc Natl Acad Sci USA

2008;105:13580-5.. A metagenomic analysis of human

microbial BSH activity.

65. Kellogg TF, Wostmann BS. Fecal neutral

steroids and bile acids from germfree

rats. J Lipid Res 1969;10:495-503

66. Kellogg TF, Knight PL, Wostmann BS.

Effect of bile acid deconjugation on the

fecal excretion of steroids. J Lipid Res

1970;11:362-6

67. Wostmann BS. Intestinal bile acids and

cholesterol absorption in the germfree

rat. J Nutr 1973;103:982-90. Investigation of cholesterol metabolism

in germ-free rats.

68. Lepercq P, Relano P, Cayuela C,

Juste C. Bifidobacterium animalis strain

DN-173 010 hydrolyses bile salts in the

gastrointestinal tract of pigs.

Scand J Gastroenterol 2004;39:1266-71

69. De Smet I, De Boever P, Verstraete W.

Cholesterol lowering in pigs through

enhanced bacterial bile salt hydrolase

activity. Br J Nutr 1998;79:185-94

70. Bateup JM, McConnell MA,

Jenkinson HF, Tannock GW.

Comparison of lactobacillus strains with

respect to bile salt hydrolase activity,

colonization of the gastrointestinal tract,

and growth rate of the murine host.

Appl Environ Microbiol 1995;61:1147-9

71. Dashkevicz MP, Feighner SD.

Development of a differential medium

for bile salt hydrolase-active Lactobacillus

spp. Appl Environ Microbiol

1989;55:11-16

72. Elkins CA, Savage DC. Identification of

genes encoding conjugated bile salt

hydrolase and transport in Lactobacillus

johnsonii100-100. J Bacteriol

1998;180:4344-9

73. Pereira DIA, McCartney AL,

Gibson GR. An in vitro study of the

probiotic potential of a

bile-salt-hydrolyzing Lactobacillus

fermentum strain, and determination of

its cholesterol-lowering properties.

Appl Environ Microbiol

2003;69:4743-52

74. Grill J-P, Schneider F, Crociani J,

Ballongue J. Purification and

characterization of conjugated bile salt

hydrolase from Bifidobacterium longum

BB536. Appl Environ Microbiol

1995;61:2577-82

75. Kim GB, Miyamoto CM, Meighen EA,

Lee BH. Cloning and characterization of

the bile salt hydrolase genes BSH from

Bifidobacterium bifidum strains.

Appl Environ Microbiol

2004;70:5603-12

76. Kim GB, Yi SH, Lee BH. Purification

and characterization of three different

types of bile salt hydrolases from

bifidobacterium strains. J Dairy Sci

2004;87:258-66

77. Tanaka H, Hashiba H, Kok J, Mierau I.

Bile salt hydrolase of Bifidobacterium

longum-biochemical and genetic

characterization. Appl Environ Microbiol

2000;66:2502-12

78. Coleman JP, Hudson LL. Cloning and

characterization of a conjugated bile acid

hydrolase gene from Clostridium

perfringens. Appl Environ Microbiol

1995;61:2514-20

79. Franz CM, Specht I, Haberer P,

Holzapfel WH. Bile salt hydrolase

activity of enterococci isolated from food:

screening and quantitative determination.

J Food Prot 2001;64(5):725-9

80. Kawamoto K, Horibe I, Uchida K.

Purification and characterization of a

new hydrolase for conjugated bile acids,

chenodeoxycholyltaurine hydrolase, from

bacteroides vulgatus. J Biochem

1989;106:1049-53

81. Stellwag EJ, Hylemon PB. Purification

and characterization of bile salt hydrolase

from Bacteroides fragilis subsp. fragilis.

Biochim Biophys Acta 1976;452:165-76

82. Kumar M, Nagpal R, Kumar R, et al.

Cholesterol-lowering probiotics as

potential biotherapeutics for metabolic

diseases. Exp Diabetes Res 2012;2012:14

83. Miyata M, Takamatsu Y, Kuribayashi H,

Yamazoe Y. Administration of ampicillin

elevates hepatic primary bile acid

synthesis through suppression of ileal

fibroblast growth factor 15 expression.

J Pharmacol Exp Ther

2009;331:1079-85

84. Kawase M, Hashimoto H, Hosoda M,

et al. Effect of administration of

fermented milk containing whey protein

concentrate to rats and healthy men on

serum lipids and blood pressure.

J Dairy Sci 2000;83:255-63

85. Andersson H, Bosaeus I, Ellegard L,

et al. Effects of low-fat milk and

fermented low-fat milk on cholesterol

absorption and excretion in ileostomy

subjects. Eur J Clin Nutr

1995;49:274-81

86. de Roos NM, Schouten G, Katan MB.

Yoghurt enriched with Lactobacillus

acidophilus does not lower blood lipids

in healthy men and women with normal

to borderline high serum cholesterol

levels. Eur J Clin Nutr 1999;53:277-80

M. L. Jones et al.

640 Expert Opin. Biol. Ther. (2013) 13(5)

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 11: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

87. Richelsen B, Kristensen K, Pedersen SB.

Long-term (6 months) effect of a new

fermented milk product on the level of

plasma lipoproteins -- a placebo-controled

and double blind study. Eur J Clin Nutr

1996;50:811-15

88. Greany KA, Bonorden MJL,

Hamilton-Reeves JM, et al. Probiotic

capsules do not lower plasma lipids in

young women and men. Eur J Clin Nutr

2007;62:232-7

89. Lewis SJ, Burmeister S. A double-blind

placebo-controlled study of the effects of

Lactobacillus acidophilus on plasma

lipids. Eur J Clin Nutr 2005;59:776-80

90. Lin SY, Ayres JW, Winkler Jr W,

Sandine WE. Lactobacillus effects on

cholesterol: in vitro and in vivo results.

J Dairy Sci 1989;72:2885-99

91. Pereira DI, Gibson GR. Effects of

consumption of probiotics and prebiotics

on serum lipid levels in humans.

Crit Rev Biochem Mol Biol

2002;37:259-81

92. Guo Z, Liu XM, Zhang QX, et al.

Influence of consumption of probiotics

on the plasma lipid profile:

a meta-analysis of randomised controlled

trials. Nutr Metab Cardiovase Dis

2011;21:844-50

93. Jeun J, Kim S, Cho SY, et al.

Hypocholesterolemic effects of

Lactobacillus plantarum KCTC3928 by

increased bile acid excretion in C57BL/

6 mice. Nutrition 2010;26:321-30

94. Taranto MP, Medici M, Perdigon G,

et al. Effect of Lactobacillus reuteri on

the prevention of hypercholesterolemia in

mice. J Dairy Sci 2000;83:401-3

95. Usman Hosono A. Effect of

administration of Lactobacillus gasseri on

serum lipids and fecal steroids in

hypercholesterolemic rats. J Dairy Sci

2000;83:1705-11

96. Begley M, Hill C, Gahan CGM. Bile

salt hydrolase activity in probiotics.

Appl Environ Microbiol

2006;72:1729-38. Review on BSH activity in

probiotic bacteria.

97. Branton WB, Jones ML,

Tomaro-Duchesneau C, et al. In vitro

characterization and safety of the

probiotic strain Lactobacillus reuteri

Cardioviva NCIMB 30242. Int J

Probiotics and Prebiotics 2011;6:1-12

98. Jones ML, Martoni C, Prakash S.

Cholesterol lowering and inhibition of

sterol absorption by Lactobacillus reuteri

NCIMB 30242: a randomized controlled

trial. Eur J Clin Nutr

2012;66(11):1234-41

99. Jones ML, Martoni CJ, Di Pietro E,

et al. Evaluation of clinical safety and

tolerance of a Lactobacillus reuteri

NCIMB 30242 supplement capsule:

a randomized control trial.

Regul Toxicol Pharmacol

2012;63:313-20. Clinical safety of BSH-active

probiotic bacteria.

100. Jones ML, Martoni CJ, Tamber S, et al.

Evaluation of safety and tolerance of

microencapsulated Lactobacillus reuteri

NCIMB 30242 in a yogurt formulation:

a randomized, placebo-controlled,

double-blind study. Food Chem Toxicol

2012;50:2216-23

101. Thomas C, Pellicciari R, Pruzanski M,

et al. Targeting bile-acid signalling for

metabolic diseases. Nat Rev Drug Discov

2008;7:678-93

102. Porez G, Prawitt J, Gross B, Staels B.

Bile acid receptors as targets for the

treatment of dyslipidemia and

cardiovascular disease. J Lipid Res

2012;53:1723-37

103. Yoon H, Ju Jh, Kim H, et al.

Lactobacillus rhamnosus BFE 5264 and

Lactobacillus plantarum NR74 promote

cholesterol excretion through the

up-regulation of ABCG5/8 in

caco-2 cells. Probiotics Antimicrobial

Proteins 2011;3:194-203

104. Yu L, Li-Hawkins J, Hammer RE, et al.

Overexpression of ABCG5 and

ABCG8 promotes biliary cholesterol

secretion and reduces fractional

absorption of dietary cholesterol.

J Clin Invest 2002;110:671-80

105. Johnson BJH, Lee JY, Pickert A,

Urbatsch IL. Bile acids stimulate ATP

hydrolysis in the purified cholesterol

transporter ABCG5/G8. Biochemistry

2010;49:3403-11

106. Huang Y, Zheng Y. The probiotic

Lactobacillus acidophilus reduces

cholesterol absorption through the

down-regulation of Niemann-Pick

C1-like 1 in Caco-2 cells. Br J Nutr

2010;103:478

107. Sudhop T, Lutjohann D, Kodal A, et al.

Inhibition of intestinal cholesterol

absorption by ezetimibe in humans.

Circulation 2002;106:1943-8

108. Altmann SW, Davis HR, Zhu LJ, et al.

Niemann-pick C1 like 1 protein is

critical for intestinal cholesterol

absorption. Science 2004;303:1201-4

109. Huang Y, Wang J, Cheng Y, Zheng Y.

The hypocholesterolaemic effects of

Lactobacillus acidophilus American type

culture collection 4356 in rats are

mediated by the down-regulation of

Niemann-Pick C1-like 1. Br J Nutr

2010;104:807-12

110. Liong MT, Shah NP. Bile salt

deconjugation ability, bile salt hydrolase

activity and cholesterol co-precipitation

ability of Lactobacilli strains. Int Dairy J

2005;15:391-8

111. Yoon Hs, Ju Jh, Kim HN, et al.

Reduction in cholesterol absorption in

Caco-2 cells through the down-regulation

of Niemann-Pick C1-like 1 by the

putative probiotic strains Lactobacillus

rhamnosus BFE5264 and Lactobacillus

plantarum NR74 from fermented foods.

Int J Food Sci Nutr

2012; [Epub ahead of print]. NPC1L1 downregulation by

probiotic bacteria.

112. Yoon Hs, Ju Jh, Kim HN, et al. The

probiotic lactobacillus rhamnosus

BFE5264 and lactobacillus plantarum

NR74 promote cholesterol efflux

and suppress inflammation in

THP-1 cells. J Sci Food Agric

2012; [Epub ahead of print]

113. Pischon T, Girman CJ, Sacks FM, et al.

Non-high-density lipoprotein cholesterol

and apolipoprotein B in the prediction of

coronary heart disease in men.

Circulation 2005;112:3375-83

114. Sniderman AD, Zhang Z, Genest J,

Cianflone K. Effects on

apoB-100 secretion and bile acid

synthesis by redirecting cholesterol efflux

from HepG2 cells. J Lipid Res

2003;44:527-32

115. Visser ME, Kastelein JJ, Stroes ES.

Apolipoprotein B synthesis inhibition:

results from clinical trials.

Curr Opin Lipidol 2010;21:319-23

116. Jones ML, Martoni CJ, Prakash S. Letter

to the editor regarding the report of

Duboc et al: connecting dysbiosis,

bile-acid dysmetabolism and gut

inflammation in inflammatory bowel

disease. Gut 2012; In press

Cholesterol lowering by bile salt hydrolase-active probiotic bacteria: mechanism, clinical evidence and heart health applications

Expert Opin. Biol. Ther. (2013) 13(5) 641

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.

Page 12: Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications

117. Danielsson H, Gustafsson B. On

serum-cholesterol levels and neutral fecal

sterols in germ-free rats. Bile acids and

steroids 59. Arch Biochem Biophys

1959;83:482-5

118. Miettinen TA, Gylling H. Regulation of

cholesterol metabolism by dietary plant

sterols. Curr Opin Lipidol 1999;10:9-14

119. Miettinen TA, Railo M, Lepantalo M,

Gylling H. Plant sterols in serum and in

atherosclerotic plaques of patients

undergoing carotid endarterectomy. J Am

Coll Cardiol 2005;45:1794-801

120. Kelly ER, Plat J, Mensink RP,

Berendschot TT. Effects of long term

plant sterol and -stanol consumption on

the retinal vasculature: a randomized

controlled trial in statin users.

Atherosclerosis 2011;214:225-30

121. AbuMweis SS, Barake R, Jones PJH.

Plant sterols/stanols as cholesterol

lowering agents: a meta-analysis of

randomized controlled trials.

Food Nutr Res 2008;52:17

122. Ortega RM, Palencia A,

Lopez-Sobaler AM. Improvement of

cholesterol levels and reduction of

cardiovascular risk via the consumption

of phytosterols. Br J Nutr

2006;96:S89-93

123. Talati R, Sobieraj DM, Makanji SS,

et al. The comparative efficacy of plant

sterols and stanols on serum lipids:

a systematic review and meta-analysis.

J Am Diet Assoc 2010;110:719-26

124. Food and Drug Administration. Food

labeling; health claim; phytosterols and

risk of coronary heart disease proposed

rules. Federal Register, Rockville,

Maryland; 2010. p. 75

125. EFSA Panel on Dietetic Products NaA.

Scientific opinion on the substantiation

of a health claim related to 3 g/day plant

sterols/stanols and lowering blood

LDL-cholesterol and reduced risk of

(coronary) heart disease pursuant to

Article 19 of Regulation (EC) No 1924/

2006. EFSA J 2012;10:2693

126. Othman RA, Moghadasian MH,

Jones PJ. Cholesterol-lowering effects of

oat B-glucan. Nutr Rev 2011;69:299-309

127. Tiwari U, Cummins E. Meta-analysis of

the effect of B-glucan intake on blood

cholesterol and glucose levels. Nutrition

2011;27:1008-16

128. EFSA Panel on Dietetic Products NaA.

Scientific opinion on the substantiation

of a health claim related to oat

beta-glucan and lowering cholesterol and

reduced risk of (coronary) heart disease

pusuant to Article 14 of regulation (EC)

No 1924/2006. EFSA J

2010;8:1885-900

129. EFSA Panel on Dietetic Products NaA.

Scientific opinion on the substantiation

of a health claim related to barley

beta-glucan and lowering cholesterol and

reduced risk of (coronary) heart disease

pursuant to Article 14 of regulation (EC)

No 1924/2006. EFSA J 2011;9:2471-87

130. Wei Z, Wang H, Chen X, et al. Time-

and dose-dependent effect of psyllium on

serum lipids in mild-to-moderate

hypercholesterolemia: a meta-analysis of

controlled clinical trials. Eur J Clin Nutr

2008;63:821-7

131. Anderson JW, Allgood LD, Lawrence A,

et al. Cholesterol-lowering effects of

psyllium intake adjunctive to diet therapy

in men and women with

hypercholesterolemia: meta-analysis of

8 controlled trials. Am J Clin Nutr

2000;71:472-9

132. McVeigh BL, Dillingham BL,

Lampe JW, Duncan AM. Effect of soy

protein varying in isoflavone content on

serum lipids in healthy young men. Am J

Clin Nutr 2006;83:244-51

133. Anderson JW, Bush HM. Soy protein

effects on serum lipoproteins: a quality

assessment and meta-analysis of

randomized, controlled studies. J Am

Coll Nutr 2011;30:79-91

134. Food and Drug Administration. Food

labeling: health claims; soy protein and

coronary heart disease. Fed Regist

1999;64:57699-733

135. Erdman JW for the AHA Nutrition

Committee. Soy protein and

cardiovascular disease. Circulation

2000;102:2555-9

AffiliationMitchell L Jones1,2,

Catherine Tomaro-Duchesneau2,

Christopher J Martoni1 & Satya Prakash†1,2

†Author for correspondence1Micropharma Ltd, 141 avenue du President

Kennedy, UQAM Biological Sciences Building,

5th Floor, Suite 5569 Montreal, Quebec,

H2X 3Y7, Canada2McGill University, Faculty of Medicine,

Departments of Biomedical Engineering,

Physiology, and Artificial Cells and Organs

Research Center, Biomedical Technology and

Cell Therapy Research Laboratory,

3775 University Street, Montreal, Quebec,

H3A 2B4, Canada

Tel: +1 514 398 3676;

Fax: +1 514 398 7461;

E-mail: [email protected]

M. L. Jones et al.

642 Expert Opin. Biol. Ther. (2013) 13(5)

Exp

ert O

pin.

Bio

l. T

her.

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

UM

EA

Uni

vers

ity L

ibra

ry o

n 08

/17/

14Fo

r pe

rson

al u

se o

nly.


Recommended