. . .. E wk... . . I‘ni 511-: a. 3M“...raas .. x, n 3. .p 3.3-5.8 v9... . .0 2 .29: . A . . |. .1 )I.. 1...... a). .- . é. , . . , . , 1.. . . .ou- I: .1- ... , , . .3... . 41v. 2 A . , Tnb’fiiéi 2&7? UBRARY Ti Michigan State UniversfiY This is to certify that the dissertation entitled INVESTIGATION OF BILE STRESS TOLERANCE AND IMMUNOMODULATORY PROPERTIES OF PROBIOTIC LACTOBACILLUS REUTERI presented by KRISTI JAMES WHITEHEAD has been accepted towards fulfillment of the requirements for the Ph.D. degree in Microbiology and Molecular Genetics M 6:54., Major Professor’s Signature “f /z=r /2 0o? l I Date MSU is an Affirmative Action/Equal Opportunity Employer PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 K‘IProlecc&Pres/ClRC/DateDue.indd INVESTIGATION OF BILE STRESS TOLERANCE AND IMMUNOMODULATORY PROPERTIES OF PROBIOTIC LACTOBACILLUS REUTERI By Kristi James Whitehead A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Microbiology & Molecular Genetics 2009 ABSTRACT INVESTIGATION OF BILE STRESS TOLERANCE AND IMMUNOMODULATORY PROPERTIES OF PROBIOTIC LACTOBACILLUS REUTERI BY Kristi James Whitehead This thesis focuses on characterization of different aspects Of L. reuteri physiology that may contribute to the probiotic potential of the species. This research includes an investigation of the bile stress response of L. reuteri ATCC 55730 (Chapter 2), analysis of the bile salt hydrolase activity of L. reuteri ATCC PTA 6475 (Chapter 3), and the investigation of the potential immunomodulatory role of lactobacillic acid, a cyclopropyl fatty acid that is specific to certain strains of L. reuteri at particular stages of growth (Chapter 4). The research contained within Chapter 2 includes an investigation of the physiological response of L. reuteri ATCC 55730 to the bile stress that would be encountered in the host small intestine, including an analysis of genes that are important for survival and growth in the presence of bile. Three genes were identified as being important for the survival of L. reuteri ATCC 55730 in bile: a putative esterase, a clp chaperone, and a gene of unknown function that is conserved within different L. reuteri isolates. One gene, encoding a multidrug resistance protein in the major facilitator superfamily, was found to contribute to the ability of the strain to grow in the presence of bile. Chapter 3 examines the ability of L. reuteri ATCC PTA 6475 to deconjugate bile acids. There is a strong correlation between strains isolated from the gastrointestinal tract and the ability to deconjugate bile acids, thus it is often suggested that bile salt hydrolase activity confers a selective advantage for colonization of the gastrointestinal tract. L. reuteri ATCC 55730, the strain investigated in the microarray and mutational analysis of the bile stress response, exhibits a very low level of bile salt hydrolase activity. L. reuteri ATCC PTA 6475, on the other hand, exhibits strong bile salt hydrolase activity against all six major human bile acids. The research contained in Chapter 3 suggests that this activity confers a growth advantage in the presence of bile acids at slightly acidic pH levels. Several strains of L. reuteri have been suggested to be able to modulate the host immune response through the production of compounds that inhibit production of the pro-inflammatory cytokine, TNF. The research in Chapter 4 identifies a possible novel immunomodulatory compound, lactobacillic acid, which may be responsible for the observed suppression. Production of this compound was demonstrated to be specific to L. reuteri strains with the TNF suppressive activity, and importantly, the appearance of lactobacillic acid in cells isolated from early stationary and late stationary phase cultures correlates with the growth stages where the immunomodulatory activity is observed. Current research isfocused on identifying the role of lactobacillic acid in TNF suppression. This work is dedicated to Daniel, who loves me unconditionally, makes me laugh, and believes in me even when I don ’t believe in myself. ACKNOWLEDGEMENTS I would first and foremost like to acknowledge my advisor, Dr. Robert Britton. I cannot thank you enough for the opportunity to work in your lab. You have been a wonderful advisor, and you have played such a huge role in my development as a scientist. Thank you so much for the patience that you have demonstrated in getting me to think for myself and in helping me to become a better public speaker. I am not sure that words can express how much the time I have spent in your lab has meant to me. I truly believe that the day that I decided to ask to join your lab was the day that I set myself on the path to becoming a successful scientist. I hope to be able to continue using the skills and lessons that l have learned from you as I continue on my scientific career. Thank you. ' The members of my advisory committee have also been an invaluable asset during my progress through graduate school. Dr. Cindy Arvidson, Dr. Lee Kroos, Dr. Thomas Schmidt, Dr. Zeynep Ustunol, and Dr. Vincent Young have all been amazing sources of knowledge and helpful discussions. Thank you all so much for your patience and guidance. To all of the past and present members of the Britton laboratory, thank you so much for making the lab a great place to work. I thoroughly enjoyed all of the debates and discussions, both scientific and non-scientific. Thank you all so much for the daily support, as well as for putting up with me during the stressful times! In particular, I would like to thank Dr. Laura Schaefer. I thoroughly enjoyed getting to know you and spend time with you in the lab and outside of the lab during my six years at Michigan State. You are a wonderful lab manager and a great friend. I would also like to acknowledge our collaborators: Jim Versalovic at Baylor College of Medicine, Stefan Roos at the Swedish University of Agricultural Sciences, and Eamonn Connolly at Biogaia. Thank you so much for lending your expertise in the field of probiotic research. You each were a wonderful source of information and guidance for many different aspects of my research. I also am indebted to Dr. Babak Borhan in the Department of Chemistry at Michigan State University and the members of his lab. This amazing group of scientists welcomed me as one of their own, both in the laboratory and during lab functions. During my time at Michigan State, I developed friendships with some truly wonderful individuals: Stephanie Eichorst, Kristin Huizinga, Jennifer Gray, Zarraz Lee, Thu Nguyen, and Adam Mosey. Without each of you to talk to, to vent to (and with), and to laugh with, I am not sure this journey would have been possible; it certainly would have been much less enjoyable. I truly hope that we can stay in contact with one another no matter where life takes us. In addition, none of this would have happened if not for the love and support that I have received both from my family and Daniel’s family. All of you have played such a strong role in the development of who I am. Thank you so much for supporting me through graduate school, especially after Daniel and I announced that we would be moving far away to Michigan. We are headed back vi closer to home now, and we are looking fonlvard to spending more time with all of you. Finally, I am forever grateful for my husband Daniel. I am not sure I believed you eleven years ago when you told me that we were meant to be together after only a few dates, but you were so confident that I decided it might be worth finding out where life took us. I could not be happier that you were right. Through high school, college, and now graduate school, you have been the absolute best friend and husband that anyone could ever ask for. You have been a constant source of love and support, always believing that I could do anything that I set my mind to. I am so proud of you and all that you have accomplished as we have progressed through the last six years of graduate school together. vii TABLE OF CONTENTS LIST OF TABLES .................................................................................. x LIST OF FIGURES ............................................................................. xii CHAPTER 1: THE PROBIOTIC POTENTIAL OF LACTOBACILLUS REUTERI INTRODUCTION .......................................................................... 1 Prevalence .................................................................................. 2 Factors that aid in colonization ......................................................... 3 Proposed mechanisms of action ..................................................... 14 Animal studies and clinical trials ..................................................... 26 REFERENCES ........................................................................... 35 CHAPTER 2: GENOMIC AND GENETIC CHARACTERIZATION OF THE BILE STRESS RESPONSE OF PROBIOTIC LACTOBACILLUS REUTERI ATCC 55730 INTRODUCTION ........................................................................ 44 MATERIALS AND METHODS ....................................................... 48 RESULTS ................................................................................. 54 DISCUSSION ............................................................................. 65 SUPPLEMENTAL INFORMATION .................................................. 72 ACKNOWLEDGEMENTS ............................................................. 82 REFERENCES ........................................................................... 83 CHAPTER 3: CHARACTERIZATION OF THE BILE SALT HYDROLASE ACTIVITY OF LACTOBA CILLUS REUTERI ATCC PTA 6475 INTRODUCTION ........................................................................ 88 MATERIALS AND METHODS ....................................................... 91 RESULTS ................................................................................. 97 DISCUSSION ........................................................................... 109 ACKNOWLEDGEMENTS ............................................................ 115 REFERENCES ......................................................................... 116 CHAPTER . 4: IDENTIFICATION OF A PUTATIVE NOVEL IMMUNOMODULATORY COMPOUND FROM LACTOBACILLUS REUTERI INTRODUCTION ....................................................................... 120 MATERIALS AND METHODS ...................................................... 123 RESULTS ................................................................................ 129 DISCUSSION ........................................................................... 145 viii ACKNOWLEDGEMENTS ............................................................ 148 REFERENCES ......................................................................... 149 CHAPTER 5: SUMMARY AND SIGNIFICANCE ...................................... 152 REFERENCES ........................................................................ 157 APPENDIX A: MICROARRAY ANALYSIS OF LACTOBACILLUS REUTERI ATCC 55730 DURING MID-LOG AND EARLY STATIONARY PHASES OF GROWTH PURPOSE ............................................................................... 159 MATERIALS AND METHODS ...................................................... 159 RESULTS ................................................................................ 161 REFERENCES ......................................................................... 171 APPENDIX B: IDENTIFICATION OF A PUTATIVE BILE-INDUCIBLE TRANSCRIPTIONAL ELEMENT FROM LACTOBACILLUS REUTERI PURPOSE ............................................................................... 172 RESULTS ................................................................................ 173 FUTURE DIRECTIONS .............................................................. 175 REFERENCES ......................................................................... 176 APPENDIX C: INVESTIGATION OF A PUTATIVE MATRIX METALLOPROTEASE FROM LACTOBACILLUS REUTERI PURPOSE ............................................................................... 177 BIOINFORMATICS .................................................................... 178 EXPERIMENTS AND RESULTS ................................................... 180 DISCUSSION ........................................................................... 184 PROTOCOLS OF INTEREST ...................................................... 187 REFERENCES ......................................................................... 196 LIST OF TABLES Table 2.1. Bacterial strains and plasmids used for this study ......................... 49 Table 2.2. Classes of genes differentially expressed during the first 15 minutes of exposure to 0.5% bile ........................................................................ 57 Table 2.3. Classes of genes differentially expressed during growth in the presence of 0.5% bile ........................................................................... 59 Table 2.4. Fold gene expression changes in the presence of bile for genes chosen for disruption ............................................................................ 61 Table 2.5. Effects of 0.3% bile exposure on cell viability and final culture density .............................................................................................. 62 Table 2.6. Genes over-expressed or under-expressed during both 15 minutes of bile exposure (0.5% oxgall) and 15 minutes of acid stress (pH 2.7) ................ 69 Table 2.7. Genes over-expressed after 15 minutes of exposure to 0.5% oxgall ................................................................................................ 72 Table 2.8. Genes over-expressed during growth in the presence of 0.5% oxgall ................................................................................................ 75 Table 2.9. Genes under-expressed after 15 minutes of exposure to 0.5% oxgall ................................................................................................ 76 Table 2.10. Genes under-expressed during growth in the presence of 0.5% oxgall ................................................................................................ 79 Table 3.1. Bacterial strains and plasmids used for this study ........................ 92 Table 3.2. Doubling times and culture densities for L. reuteri ATCC PTA 6475 and PRBZ40 in the presence and absence of human bile acids ................... 104 Table 3.3. Observed growth effects of individual bile acids in plates or in liquid medium on L. reuteri ATCC PTA 6475, PR8240, and L. reuteri ATCC 55730. Notable differences between ATCC PTA 6475 and PRBZ40 are highlighted in bold. ND = not determined. *Concentrations of greater than 0.1% were not tested for the deconjugated bile acids due to solubility problems .................. 107 Table 4.1. Bacterial strains and plasmids used in this study ........................ 124 Table A1. Genes significantly over-expressed by L. reuteri ATCC 55730 during early stationary phase ......................................................................... 163 Table A2. Genes significantly under-expressed by L. reuteri ATCC 55730 during early stationary phase ......................................................................... 167 I i xi LIST OF FIGURES Figure 2.1. Representative growth curve of L. reuteri ATCC 55730 used for microarray experiments (arrows represent time points where samples were taken for RNA isolation). Open arrows represent samples for bile shock experiments; filled arrows represent samples for bile adaptation experiments. 0.5% oxgall was added at 250 minutes ........................................................................... 52 Figure 2.2. Comparison of survival after 30 minutes of exposure to 0.3% oxgall for L. reuteri ATCC 55730 wild-type and PRB190 (lr1864 - CIpL), PRB188 (lr1516 - putative esterase), PRB167 (er085 - unknown), PRB114 (Ir1706 - Dps), PRB126 (Ir1265 - multidrug resistance protein in the ABC transporter family), and PRB130 (Ir1584 - multidrug resistance protein in the major facilitator superfamily) mutant strains. Cultures were plated onto MRS plates after bile exposure to determine the number of viable cells. Error bars represent standard deviation. *p < 0.001 compared with wild-type ......................................................... 63 Figure 3.1. Different phenotypes observed using the plate test to determine bile salt hydrolase activity. Strains of L. reuteri were plated onto MRS plates with or without bile acids to determine bile salt hydrolase ability. A. L. reuteri ATCC PTA 6475 and the bile salt hydrolase mutant strain, PRBZ40, plated onto MRS plates containing 0.5% oxgall. Precipitation of bile is observed from ATCC PTA 6475 as halos around the colonies; no activity is observed for PR8240. This phenotype was also observed for plates containing glycodeoxycholate (GDCA) and glycochenodeoxycholate (GCDCA) (data not shown). B. The two strains (ATCC PTA 6475 and PR8240) plated onto MRS plates containing 0.01% taurochenodeoxycholate (TCDCA). Bile salt hydrolase activity for this bile acid was demonstrated by a cloudiness surrounding the colonies that could be observed when the plates were held against a light. No activity is observed for PR8240. C. ATCC PTA 6475 plated onto MRS or MRS plus 0.5% taurodeoxycholate (TDCA). Deconjugation of TDCA was demonstrated by a matte opaque granular appearance of the colonies. This phenotype was not observed for PR8240 (data not shown) .................................................... 99 Figure 3.2. Deconjugated bile acids begin to accumulate after 4 hours in a culture of L. reuteri ATCC PTA 6475. Representative TLC plate showing the bile salt hydrolase activity of L. reuten' ATCC PTA 6475 against a synthetic “human” bile acid mixture (SHB). Samples were taken a 1, 4, 7, and 23 hours. Control solutions of cholate (CA) and deoxycholate (DCA) are included for comparison [deoxycholate and chenodeoxycholate (CDCA) migrate at the same rate in this solvent system]. The bile salt hydrolase mutant strain, PRB240, is shown to have no activity at the 23-hour time point ................................................. 103 xii Figure 3.3. Controlling the pH of the culture does not affect the bile salt hydrolase activity of L. reuteri ATCC PTA 6475. Representative TLC plate showing the bile salt hydrolase activity of L. reuteri ATCC PTA 6475 against a synthetic “human” bile acid mixture (SHB) in MRS and buffered MRS. Samples were taken at 7 hours. Control solutions of cholate (CA) and deoxycholate (DCA) are included for comparison [deoxycholate and chenodeoxycholate (CDCA) migrate at the same rate in this solvent system]. No difference in activity is observed between cultures grown in MRS and cultures grown in buffered MRS ................................................................................................ 106 Figure 4.1. Synthesis of cyclopropane fatty acids. Cyclopropane fatty acid synthase converts oleic acid and vaccenic acid into dihydrosterculic acid and lactobacillic acid, respectively ............................................................... 122 Figure 4.2. A representative growth curve for L. reuteri ATCC PTA 6475. Samples for fatty acid analysis (mid-log, early stationary, and late stationary phase) were collected at the time points represented by open circles ............ 127 Figure 4.3 - TNF-inhibitory strains produce lactobacillic acid. Cultures of five strains of L. reuten' were grown in MRS broth for 18 hours at 37° C. Fatty acid profiles were determined by gas chromatography. ATCC 55730 and CF48-3A (TNF non-inhibitory strains) do not produce the CFA, lactobacillic acid. In contrast TNF-inhibitory strains (ATCC PTA 6475, ATCC PTA 5289, and ATCC PTA 4659) produce lactobacillic acid. Experiments were performed in triplicate; error bars represent standard deviation. *Lactobacillic acid ............................................................... 132 Figure 4.4. Lactobacillic acid appears in late stationary phase cultures of L. reuten' ATCC PTA 6475. Cultures of L. reuteri ATCC PTA 6475 were grown in MRS broth at 37°C. Samples were collected for FAME analysis at different stages of growth. Experiments were performed in triplicate; error bars represent standard deviation. *Lactobacillic acid ........................ 134 Figure 4.5. Disruption of NT01LR1143 (CFA synthase) eliminates production of lactobacillic acid by L. reuteri ATCC PTA 6475. L. reuteri ATCC PTA 6475 and PRB173 were grown overnight in MRS broth at 37° C. Samples were collected for fatty acid analysis. Experiments were performed in triplicate; error bars represent standard deviation ................................................................. 136 Figure 4.6. Lactobacillic acid production by L. reuteri ATCC PTA 6475 can be increased by removal of Tween 80 from the growth medium. Overnight cultures of L. reuteri ATCC PTA 6475 were grown in MRS A (complete MRS broth), MRS B (MRS broth with Tween 80 and beef extract removed), or MRS C (MRS B plus 0.01 % cis-vaccenic acid); samples were collected for fatty acid analysis. xiii Experiments were performed in triplicate; error bars represent standard deviation ........................................................................................... 139 Figure 4.7. An insertion in the CFA synthase gene of L. reuteri ATCC 55730 and CF48-3A may prevent production of lactobacillic acid. PCR analysis revealed an insertion in the region of the genome containing the cfa gene for both immunomodulatory strains. This insertion results in the truncation of the cfa gene. Small arrows represent the primers used for amplification of the region surrounding the cfa gene ..................................................................... 141 Figure 4.8 - L. reuteri deficient in Cfa had diminished ability to suppress TNF. Cell-free supernatants from L. reuten' cultured as planktonic cells or biofilms were tested for the ability to inhibit LPS-activated THP-1 cells from producing TNF. When cultured as planktonic cells (A) or biofilms (B), PBR173, which does not produce lactobacillic acid, had diminished ability to suppress TNF production compared to wild-type cells .................................................................. 144 Figure A.1. A representative growth curve of L. reuteri ATCC 55730. Samples for microarray analysis (mid-log and early stationary phase) were taken at the time point represented by the open circles ............................................... 162 Figure 8.1. The sequences of the 13 occurrences of the possible bile-inducible transcriptional element in L. reuteri ATCC 55730 identified by MEME ............ 174 Figure C.1. The protein sequence of L. reuteri ATCC PTA 6475 NT01LR0595 (Lr1291 homolog). The putative cleavage site is marked with two forward slashes (ll); this site was predicted by SignalP 3.0. The putative active site is underlined, with the conserved residues in bold. The conserved methionine residue (“met-turn”) is also in bold .......................................................... 179 xiv CHAPTER 1 THE PROBIOTIC POTENTIAL OF LACTOBACILLUS REUTERI Introduction and. ' Probiotics, which are defined as “live microorganisms which when administered in adequate amounts confer a health benefit on the host" (19), have become increasingly popular over the past several decades as reports of antibiotic resistant infections and the prevalence of gastrointestinal disorders such as inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS) continues to rise. Despite the increase in usage, not much is known about the mechanisms through which these microorganisms may be able to cause their beneficial effects. Production of antimicrobial compounds, alterations of the host microbiota, and modulation of the immune response of the host are among the proposed mechanisms (18, 54, 62). In part because the mechanisms through which these microbes cause their beneficial effects are not known, the characteristics that make an effective probiotic are also speculative. Some general characteristics that are proposed to be important include that the microbe be of host origin, is nonpathogenic, is able to resist the stresses encountered during passage through the host, and is able to confer a beneficial effect on the host (18). Lactobacilli are among the most commonly used bacteria in probiotic products, in part due to their long history of safe use in the food industry. Among the lactobacilli, many different species are currently being investigated as effective probiotics, including various strains of L. acidophilius, L. rhamnosus, and L. reuteri (17). L. reuteri is a heterofermentive species of lactic acid bacteria that is found natively in the gastrointestinal tracts of humans (52). This review will summarize the recent literature regarding the potential of various strains of L. reuteri to be used as effective probiotics; both in vitro experiments and clinical trials will be discussed. Prevalence Although there is a lack Of knowledge regarding what characteristics make an effective probiotic, it is generally considered to be important for the bacteria to have been isolated from the host in which it will be used. Lactobacillus reuteri is one of the few species of lactic acid bacteria that is naturally found in humans as well as in a variety of animals as part of the normal microbiota. Using culture- based isolation techniques, L. reuteri has been found to be one of the dominant species of Lactobacillus isolated from chickens, mice, dogs, and pigs (2, 39, 41, 48, 79, 89). Within the human gastrointestinal tract, only a handful of lactic acid bacteria are considered to be autochthonous; these include L. reuteri, L. gasseri, and possibly L. ruminus and L. salivarius (52). Although bacterial samples are often taken from the intestines or fecal samples for analysis, there are several reports that suggest that L. reuteri may also inhabit the stomach of humans (52, 57, 78), a location typically considered to be fairly uninhabitable for bacteria due to the extremely low pH. In a recent 28-day clinical trial, Valeur et al (78) administered L. reuteri ATCC 55730, a probiotic strain of human-origin, to participants. They were able to demonstrate that the strain survived in the stomach, duodenum, and ileum of the study participants. This supports the argument that strains isolated from the host are most likely already adapted for survival in the host, and therefore, may be strains worth investigating for probiotic usage. Factors that aid in colonization. Although being of host origin is generally considered to be an important trait, many of the potential probiotic strains currently in use originated from the food industry. As the definition of a probiotic requires the organism to survive passage through the gastrointestinal tract, there are several characteristics that are generally investigated through in vitro assays before the strains are used in animal studies or clinical trials. These traits include resistance to stresses encountered in the gastrointestinal tract (such as low pH in the stomach and bile in the small intestine), as well as the ability to attach to host surfaces for colonization. It is also generally desirable to understand how strains are able to survive these harsh conditions. Knowledge of the mechanisms could allow researchers to quickly screen for strains that can survive passage through the gastrointestinal tract based on a particular trait or allow for improvement of the survival rate of a strain with intriguing beneficial properties that may have poor survival or colonization abilities. As multiple strains of L. reuteri have been shown to survive in the gastrointestinal tract in a wide variety of animals, this species may be ideal for investigating these mechanisms. Acid stress. As previously mentioned, several studies have shown that L. reuten' can survive in the human stomach (52, 57, 78), an environment generally considered to be inhospitable to bacteria due to its extremely low pH. The pH in the human stomach can reach as low as 1.5 during the fasting stage, with values ranging between 3.0 to 5.0 after consumption of a meal (16). One of the most commonly used delivery mechanisms for probiotics, yogurt and other fermented dairy products, are also typically acidic (16). Therefore, an understanding of the ways in which various strains of L. reuteri can survive lOw pH exposure is beneficial for host, as well as industrial, applications. Lee et al (37) exposed L. reuteri ATCC 23272 to pHs of 5.0, 4.5, and 4.0 for one hour and compared the protein expression under these conditions to cultures grown at pH 6.8. They identified 40 proteins involved in a wide range of cellular functions that were significantly differentially expressed during incubation at all three lower pH values, suggesting that L. reuteri has a complex response to pH stress. Proteins over-expressed during growth at lower pHs included those involVed in transport and binding, transcription and translation, metabolism, stress, and pH homeostasis. In a more focused study, microarray and mutational analyses of the acid shock response of L. reuteri ATCC 55730 were performed by Wall et al (81 ). The gene expression changes of cells that were exposed to pH 2.7 for either 5 or 15 minutes were identified based on comparison with the gene expression profiles of cells grown at pH 5.1. Overall, 72 genes were found to be differentially expressed when L. reuteri ATCC 55730 was exposed to acid shock conditions. Viability studies at pH 2.7 revealed that for this strain, more than 80% of the cells survived exposure for one hour, although no growth appeared to take place. This suggests that although the strain is able to survive passage through the stomach and even possibly colonize the stomach (78), time may be needed to adapt to the harsh acidic conditions before growth can resume. Such an adaptation period lasting approximately two hours was demonstrated when L. reuteri ATCC 55730 was exposed to bile in vitro (87). Based on the microarray analysis, two genes were chosen for disruption; lr1864 (clpL, a putative ATPase that may act as a chaperone) and lr1516 (a putative esterase in the penicillin binding protein family). Both of these mutants were found to have significantly lower survival rates compared to the wild-type strain when incubated in a synthetic gastric juice at pH 2.0 for 50 minutes. The mutational analysis suggests that the CIpL chaperone may be needed to aid the cells in degrading or refolding proteins denatured by the low pH exposure; it also suggests that the putative esterase may be involved in altering the cell wall in such a way that allows the cells to survive exposure to low pH (81). Further study is needed to fully understand how L. reuteri responds to and survives exposure to the low pH found in the stomach. The importance of CIpL and the putative esterase should be investigated in an in vivo model to determine if they truly contribute to survival in the gastrointestinal tract. Bile stress. After exposure to the low pH in the stomach, probiotic bacteria then move into the small intestine where they are exposed to bile, a complex mixture that typically aids in fat digestion and absorption (27). Bile acids, the main component in bile, are known to have potent antimicrobial effects (25), yet as previously mentioned, some strains of L. reuteri have been shown to colonize portions of the small intestine (78). The response of these strains to bile is important for understanding what physiological changes the bacteria undergo during passage through the gastrointestinal tract. Bile acids are amphipathic molecules that function to solubilize fats. Their amphipathic nature allows them to form mixed micelles with fats and cholesterol in the body and also allows them to act as detergents. One of the main ways through which bile is thought tO cause damage to bacterial cells is through disruption of the cell membrane and wall. Taranto et al (71) demonstrated that growth in the presence of bile induced changes in the cell membrane of L. reuteri CRL 1098. Exposure to bile decreased the amount of phospholipids and also decreased the ratio of saturated to unsaturated fatty acids in the cell membrane. These changes may be important for survival during passage through the gastrointestinal tract. L. reuteri ATCC 55730 was shown to survive in physiologically relevant concentrations of bile in vitro (87). Growth studies revealed that there was an initial pause in growth when mid-log phase cells were exposed to bile. This pause in growth is believed to be a period of adaptation, during which the cells make physiological changes that allow them to then grow in the presence of bile. In order to determine how bile exposure affects the gene expression of L. reuteri and to understand what changes allow the cells to resume growth in the presence of bile, microarray analyses were performed comparing the gene expression profiles of cells before and after bile exposure. Based on the microarray analysis, several genes were chosen for mutational analysis. Disruptions in three genes were shown to cause significantly lower survival rates during the first 30 minutes of bile exposure. These genes included lr1864 (clpL), lr0085 (a gene of unknown function that is conserved among L. reuteri only), and lr1516 (a putative esterase). In addition, a disruption in another gene, lr1584 (a multidrug resistance protein in the major facilitator superfamily), appeared to affect the ability of the cells to grow in the presence of bile, although the initial survival rate was not altered (87). The growth defect observed in the lr1584 mutant is in accordance with known mechanisms of bile resistance for gram- negative organisms, where the utilization of efflux pumps to expel bile acids has been demonstrated (25). Interestingly, there is overlap in the gene expression changes of L. reuteri exposed to low pH and bile based on microarray analysis (81, 87). There is also overlap in the mutational analysis; ClpL (lr1864) and the putative esterase (lr1516) were both shown to be important for survival in low pH and bile. This overlap suggests that exposure to acid in the stomach may prepare L. reuteri for Anna--1." exposure to bile in the small intestine. Further research is required to determine if the period of adaptation observed in vitro during bile exposure may be decreased by a prior exposure to low pH. One other possibility of how L. reuteri may deal with the stress of bile acids is through the activity of a bile salt hydrolase (BSH) enzyme. When bile is secreted into the small intestine from the gall bladder, the majority of bile acids are conjugated to either a glycine or taurine molecule; however, by the time the bile reaches the end of the small intestine, almost all of the bile acids are unconjugated (i.e. the glycine or taurine has been removed) (47). This deconjugation activity is due to bacterial bile salt hydrolase enzymes and plays a role in bile acid metabolism of the host (7). There is much debate as to the role of the bacterial BSH enzymes. Some possible roles include lowering the cholesterol of the host, providing a nutritional benefit to the bacteria, or detoxification of the bile acids to protect the bacteria (7). Taranto et al (74) demonstrated that L. reuteri CRL 1098 contained an intracellular bile salt hydrolase enzyme that was expressed when the cells were in stationary phase. Whether or not this enzyme plays a role in bile resistance of L. reuteri is yet to be determined. Research in our lab suggests that there is substantial strain variation in BSH enzyme specificity and activity among various L. reuteri (unpublished observation). Attachment. It has been suggested that probiotic bacteria should persist in the gastrointestinal tract in order to cause their beneficial effects; the ability to attach to intestinal surfaces such as epithelial cells or mucus is often proposed to aid in this persistence. Several studies have examined the ability of L. reuteri to adhere to mucosal surfaces using cell lines, mucus components, or animal models. Wang et al (85) tested nine different strains of Lactobacillus for their ability to attach to human enterocyte-like HT-29 cells, as well as to porcine gastric mucin, one of the main components of intestinal mucus. Of the nine strains, they found that L. reuteri JCM1081, a chicken isolate, was best able to adhere. Further analysis identified a 29 kDa protein, a putative ATP-binding cassette transporter protein, that was able to bind to both the cell line and the mucin in vitro, suggesting that this protein may be responsible for the adherence observed. The authors propose that based on these experiments mucin may actually act as a receptor for the binding of probiotic bacteria in the gastrointestinal tract. Roos et al (55) also previously identified a 29 kDa collagen binding protein from L. reuteri NCIP 11951 similar to ABC-type transport proteins that was suggested to aid in attachment of the bacterium to epithelial surfaces. Sequence comparison revealed these two proteins to be similar (85), suggesting a conserved binding protein between the two strains. Although there may be conserved traits identified, strain specificity is something that should be taken into careful consideration when investigating L. reuteri for probiotic usage. Characteristics that may be important for causing beneficial effects, such as binding to intestinal surfaces, may vary substantially between strains. For example, Miyoshi et al (42) tested L. reuteri strains from various sources (human, pig, mouse, and chicken) for their ability to adhere to Caco-2 cells in vitro. They identified three strains that had high levels of adherence (L. reuteri DSMZ 20016, 104R, and LEM83) and two strains that had low levels of adherence (L. reuteri ML1 and LB54). Upon further investigation, they identified a protein, MapA (mucus adhesion promoting protein) that also bound to Caco-2 cells and had the ability to inhibit the binding of L. reuteri to Caco-2 cells in a concentration-dependent manner. Although MapA appears to be a protein that is involved in binding to eukaryotic cells, all strains utilized in this study were found to contain the gene for MapA, despite the different adherence levels. Also, although MapA was able to compete with L. reuteri for binding, approximately 50% of the bacterial cells were able to bind to Caco—2 cells, even under conditions using saturating amounts of MapA. These observations suggest that binding to Caco-2 cells involves more than one factor, and that binding activity most likely is controlled by different factors in different strains of L. reuteri. 10 Although some studies have investigated binding to both cells and mucus or mucus components, other studies have focused singularly on the ability of L. reuteri to bind to mucus, since the epithelial surface in the gastrointestinal tract is covered with a mucus layer in healthy individuals. Jonsson et al (31) investigated the ability of seven different strains of L. reuteri to bind to pig gastric mucin. They observed varying levels of attachment between the strains. When the strains were grown in the presence of mucin, an increase in adherence was observed for strains with previously low levels of binding, whereas an inhibition of binding was observed for strains with naturally high levels of adherence. Proteinase K treatment was found to abolish binding, suggesting that binding is due to a protein. The effects observed by growth of the strains in the presence of mucin could also be explained by the dependence of binding on a protein or proteins. Growth in mucin could induce expression of a protein necessary for binding in the strains with naturally low levels of binding, but the presence of mucin could compete with a binding protein already present in the strains with naturally high levels of binding. This binding is not believed to be due to the previously identified 358 kDa cell surface protein, Mub, which was identified from L. reuteri 1063 and shown to bind to mucus and mucus components in vitro (56), as only one of the seven strains included in this study is known to produce Mub (31). 11 Although in vitro adherence tests provide a rapid means of examining the potential of certain strains to persist in the intestine, little is known about how host factors and physiological changes that the bacteria undergo during passage through the host may affect persistence. Initial studies in the host have identified a wide range of factors that can affect colonization success, including various cell surface proteins, exopolysaccharide production, and D-alanylation of teichoic acids in the cell membrane and wall. Walter et al (82) examined the effect of disruption of four genes on the survival of L. reuteri 100-23 in the mouse. They identified one mutant with a disruption in Lsp (a high-molecular—mass surface protein) that was delayed in its colonization rate and which had impaired adherence to the forestomach epithelium of the mouse. This mutant was also found exhibit lower colonization levels when inoculated in competition with the wild-type strain. These experiments suggest that cell surface proteins are important for adherence and colonization in vivo, supporting the previously described in vitro data. The study also demonstrated somewhat reduced performance in an MsrB mutant (methionine sulfoxide reductase), although there was large inter-animal variability for this mutant. Exopolysaccharide (EPS) production may also play a role in persistence for some strains. In L. reuteri TMW1.106, two proteins involved in EPS production were found to affect colonization of the strain in the gastrointestinal tract of mice. thA (glucosyltranferase) and lnu (inulosucrase) mutants were both found to be 12 defective in colonization of the mouse. The inu mutant was defective in colonization when put into competition with the wild-type strain; the 9th mutant was defective in colonization when it was inoculated in competition with L. johnsonii #21, but not when it was inoculated in competition with the wild-type L. reuteri, suggesting that the wild-type strain was somehow able to complement the observed defect in the mutant (84). Finally, the effect of D-alanylation of teichoic acids in the bacterial cell membrane and wall has been investigated in L. reuteri. The incorporation of D-alanyl esters into teichoic acids has been shown to be important for bacterial virulence when studied in various pathogens (1, 15, 51). In order to investigate whether there is overlap between the methods that pathogens and beneficial bacteria use to colonize the host, Walter et al (83) disrupted the D-alanylation operon of L. reuten' 100-23 by insertional disruption of dltA, the second gene in this operon. The dltA mutant was defective for colonization of the mouse when inoculated in competition with the wild-type strain, as well as when it was inoculated alone. The mutant also was demonstrated to be defective in the ability to form a biofilm on the murine forestomach epithelium. In vitro studies shoWed that the mutant had impaired growth under acidic conditions, which may play a role in the observed in vivo defects. Overall, attachment to mucosal surfaces is a complex phenomenon that appears to be dependent on multiple bacterial surface characteristics such as EPS 13 production and cell surface proteins. Although there may be some conservation of characteristics important for attachment and persistence, there also appears to be considerable strain variation with L. reuteri that should be taken into consideration. Proposed Mechanisms of Action Although the popularity of probiotics has increased over the last few decades and they are being investigated as possible treatments for a wide variety of ailments, the mechanisms of action for the beneficial effects observed with probiotic treatment are still unknown. Some of the proposed mechanisms of action include pathogen inhibition, primarily through the production of antimicrobial compounds, and modulation of the host immune response (54, 62). Various strains of L. reuteri have been investigated for these traits. Pathogen inhibition - production of antimicrobial compounds. Although production of hydrogen peroxide and lactic acid are commonly thought to contribute to pathogen inhibition by lactic acid bacteria (34, 35), most of the research regarding pathogen inhibition by L. reuteri has focused on other antimicrobial factors. L. reuteri is known to produce at least three other separate antimicrobial compounds: reuterin, a by—product of glycerol metabolism, reutericylcin, a novel tetramic acid derivative, and reutericin, a bacteriocin. 14 Reuterin. One of the most widely studied antimicrobial compounds produced by L. reuteri, and the reason commonly attributed for the beneficial effects observed with administration of the species, is reuterin, a by-product of glycerol metabolism. Reuterin has been identified as an equilibrium mixture of 3- hydroxypropionaldehyde (3-HPA) in the monomer form, the hydrate form, and the cyclical dimer form (70). Reuterin has been found to have a wide range of antimicrobial activity, including antibacterial, antimycotic, and antiprotozoal activities (3, 13). Lactobacilli are generally more resistant to reuterin than other microbes; one study found that at least two times the reuterin was needed to kill lactic acid bacteria as compared to other bacteria (13, 14). Although other organisms have been shown to produce 3-HPA as a by-product of glycerol metabolism (5, 20, 21, 40, 60), L. reuteri appears to be fairly unique in the ability to accumulate large amounts of reuterin (although this trait is possibly shared with certain strains of L. coryniformis (40)). A recent study demonstrated that L. reuteri had the highest reuterin Mle (minimum inhibitory concentrations) when compared to other intestinal bacteria (14). Recent work has focused on the possible mechanism of action for reuterin’s antimicrobial effects, as well as on attempts at determining whether reuterin is a relevant antimicrobial compound in vivo. Morita et al (44) were able to show that reuterin can be produced in detectable amounts in vivo using gnotobiotic BALB/c mice monoassociated with L. reuteri JCM 1112 if glycerol is present. Their research also demonstrated that 7 to 10 mM glycerol was found in the fecal 15 material of untreated mice, suggesting that reuterin may be produced in concentrations relevant for antimicrobial activity in vivo. Schaefer et al (63) have suggested that reuterin causes its broad-spectrum antimicrobial effects by triggering oxidative stress through modification of thiol groups. This mechanism is supported by an overlap in the gene expression profile of Escherichia coli cells treated with reuterin and the known OxyR regulon, a group of genes known to be involved in responding to various oxidative stresses. The modification of thiol groups suggests that the aldehyde form of 3- HPA is most likely the active portion of the equilibrium mixture (63). One intriguing characteristic about reuterin production is that larger amounts of reuterin are accumulated in the media when other bacteria are present. Various bacteria have been shown to cause this increased accumulation of reuterin (13, 63). Schaefer et aI recently observed that live bacteria and UV-killed bacteria but not heat—killed bacteria, triggered this increased accumulation (63). It has not been determined whether the excess accumulation is due to increased reuterin production or increased secretion of reuterin. Although reuterin is commonly attributed as one of the main reasons for the observed beneficial effects with L. reuteri, care should be taken to confirm this as the active trait for individual strains and in specific instances. Despite the evidence that reuterin is produced by various strains of L. reuteri, accumulation 16 levels can differ (69), and some strains have been shown to not contain the necessary enzymes for reuterin production (8). Some research also suggests that the genes necessary for reuterin production may have been horizontally acquired (44), possibly explaining the lack of these enzymes in certain strains. L. reuteri RC-14, a strain demonstrated to have many beneficial effects in urogenital health, does not make reuterin and does not contain the glycerol dehydratase enzyme necessary for converting glycerol into 3-HPA. Therefore, any probiotic effects observed with this strain are not due to reuterin production (8), and other possible mechanisms of action should be explored. Reutericyclin. Reutericyclin was first identified as a low-molecular weight antimicrobial compound that is produced by L. reuten' LTH2584, a strain isolated from sourdough bread fermentations (22), although more recent research has demonstrated that it is produced by several different strains isolated from sourdough fermentations (23). Holtzel et al (29) identified reutericyclin as a novel tetramic acid derivative, 3-acetyl-1-(2-trans-decenoyl)-2-hydroxy-(5R)-isobutyl-A2- pyrroline-4-one. Reutericyclin has been shown to have a broad-spectrum antimicrobial activity against various gram-positive organisms, including Lactobacillus spp., Bacillus subtilis, Bacillus cereus, Enterococcus faecalis, Staphylococcus aureus, and Listeria innocua (22). Ganzle et al (24) demonstrated that the antimicrobial activity observed for reutericyclin was due to its ability to dissipate the transmembrane proton potential of sensitive cells. The main role of reutericyclin has been proposed as contributing to the persistence of 17 L. reuteri in sourdough fermentations. This proposed role is due to exhibited production of active amounts of reutericyclin in sourdough fermentations by L. reuteri (23), although due to the wide-spectrum inhibitory activity of the compound against gram-positive bacteria, further investigation of this compound is warranted. Reutericin. Bacteriocins are antimicrobial proteinaceous compounds that have activity against closely related bacteria (53). At least one strain of L. reuteri, L. reuteri LA6, is known to produce a bacteriocin, reutericin 6. Reutericin 6 is a 2.7 kDa protein (32) that has been shown to have bacteriocidal activity against other lactobacilli, including L. acidophilus and L. delbruckii. The role of reutericin 6 has been proposed as a way for L. reuteri to remain the dominant species of heterofermentive lactobacilli in the gut (75). Reutericin 6 and gassericin A (a bacteriocin produced by Lactobacillus gasseri LA39) have been demonstrated to be identical in their molecular weight and primary amino acid structure, yet vary in the spectrum of their antimicrobial activity. The proposed reason for this difference in activity is a difference in the D-amino acid content of the two proteins (33). Investigation into the specificities of these two compounds is needed to determine the role of these similar compounds that are both produced by bacteria isolated from the same location, the feces of a human infant (33). Further research into whether these two organisms use the compounds to compete or to cooperate could indicate whether a mixture of these two strains might be advantageous for probiotic usage. 18 Pathogen inhibition - displacement. Apart from actually inhibiting the growth of or killing various pathogens, it is also possible that probiotics such as L. reuteri exert their beneficial effects through competition for binding sites or nutrients. Various studies have investigated the ability of L. reuteri to either inhibit the attachment of pathogens or to displace pathogens that have already adhered. Strain specificity appears to have a large effect on the ability of L. reuteri to inhibit binding of pathogens to epithelial cells. Vesterlund et al (80) found that L. reuteri ING1 not only had significantly lower adherence levels to mucus than Staphylococcus aureus, but the strain also was not able to displace adherent S. aureus, unlike other lactic acid bacteria tested in the study. They were, however, able to demonstrate a significant reduction in the viability of S. aureus when incubated with L. reuteri ING1 in the presence of 1% glycerol, suggesting that this strain may still be effective in preventing infection by S. aureus. It is also important to consider the effects that certain probiotic strains may have on one another, since many probiotic companies are marketing products containing mixtures of various strains. Larsen et al (35) tested multiple strains of Lactobacillus, including three strains of L. reuteri for the ability to bind to porcine epithelial cells and to either inhibit binding of or promote displacement of other lactobacilli or pathogenic E. coli 0138 using in vitro assays. The authors of this study not only demonstrated a wide range in the adherence capabilities of the 19 three L. reuteri strains (ranging from 3.5 to 38% of bacteria), but also found that the most strongly adherent strain, L. reuteri DSM 12246, could actually displace other lactobacilli. Interestingly, in this study, there appeared to be no correlation between the ability of the lactobacilli to adhere and the inhibition of the E. coli 0138, as all tested lactobacilli reduced adherence of the E. coli (35). One possibility for inhibition of binding or displacement of pathogens is through competition for binding sites. Todoriki et al (76) tested various strains of lactobacilli for the ability to adhere to Caco-2 cells in vitro. They demonstrated that a strain of L. reuteri, JCM 1081, had the highest level of adhesion, while L. crispatus JCM 8779 had the second highest level of adhesion. Both of these strains were able to lower adherence levels Of E. coli, Salmonella typhimun'um, and Enterococcus faecalis. Although the adherence inhibition was suggested to be due to antimicrobial activity in the L. crispatus culture, no such activity was observed with the L. reuteri culture, suggesting either competition for binding sites or steric hindrance was the cause of the observed adherence inhibition. Using a more specific example, out of nine L. reuteri strains tested, two were shown to not only bind to both putative glycolipid receptor molecules of Helicobacter pylori, but also to inhibit binding of H. pylori to both, suggesting a competition for receptor binding (45). Another interesting connection regarding competition for binding sites is that Heinemann et al (26) identified a 29 kDa protein (as part of a biosurfactant mixture) from L. reuteri RC-14 that has strong anti-adhesive properties against Enterococcus faecalis 1131. The authors 20 suggest that this protein is the same one identified by Roos et al (55) that is believed to play a role in adherence of L. reuteri NCIB 11951. The identification of the same protein during studies investigating adhesion of a probiotic strain to host tissues and protection of host tissues by a probiotic strain suggest there may be similarities in the adherence mechanisms of probiotic and pathogenic strains which could lead to competition for binding sites. One study has shown that L. reuteri may not only be able to displace individually bound bacteria, but also can have an effect on bacterial communities such as biofilms. L. reuteri RC-14, a strain demonstrated to have beneficial effects on urogenital health, was tested to determine its effect on biofilms formed by Gardnerella vagina/is, a pathogen commonly associated with bacterial vaginosis. L. reuteri RC-14 was found to disrupt and invade G. vagina/is biofilms in vitro, as well as cause a significant reduction in viability of the pathogen after co- incubation for 24 hours (59), suggesting that this strain can have a detrimental effect on an already well-established pathogen. Pathogen inhibition - alteration of virulence factor expression. One other possibility for probiotic benefit through direct inhibition of pathogens is the possibility that the probiotic bacteria are able to alter either gene or protein expression of the pathogen’s virulence factors. There are several in vitro studies that suggest that this may be yet another possible mechanism of action for L. reuteri. 21 Several studies have investigated the response of E. coli O157:H7 to incubation with L. reuteri. Carey et al (11) demonstrated that various strains of Bifidobacterium, Pediococcus, and Lactobacillus, including L. reuteri were able to decrease expression of Stx2A, the gene that encodes the A subunit of Shiga toxin 2. A different study demonstrated that culture supernatants from a certain strain of L. reuteri, L. reuteri ATCC 55730, were able to repress E. coli O157:H7 ler expression. The main virulence factors for E. coli O157:H7 are found in a pathogenicity island termed the locus of enterocyte effacement or LEE. The regulator of expression for this pathogenicity island is ler (or LEE regulator). The ability of L. reuteri culture supernatants to suppress expression of ler was found to be strain dependent, as supernatants from L. reuteri ATCC 55730 repressed expression, but supernatants from L. reuten' 100-23 actually induced expression of Ier (30). Protein expression from pathogens can also be affected, as evidenced by co- incubation of Staphylococcus aureus with L. reuteri RC-14 (36). Research demonstrated that secreted molecules of L. reuteri RC-14 were able to trigger a decrease in expression of SSL11 (staphylococcal superantigen-Iike protein 11) by S. aureus. SSL11 is a putative exotoxin that is proposed to be a virulence factor for S. aureus. The authors also demonstrated that the secreted molecules responsible for the suppression were not sensitive to protease treatment, suggesting that proteins are not responsible for the observed effects. The 22 responsible secreted molecules were shown to repress the SSL11 and P3 promoters of S. aureus, further suggesting that L. reuteri RC-14 may be able to alter the virulence of this common pathogen (36). Immunomodulation. Apart from the potential for probiotic strains to cause their beneficial effects by either altering the microbial community of the host through production of antimicrobial compounds or through the prevention of establishment of various pathogens, one other popular proposed mechanism of action is the ability of probiotic bacteria to modulate the immune response of the host. Various studies have been performed to investigate the effect that administration of L. reuteri may have on dendritic cell maturation and cytokine production. Dendritic cells are antigen-presenting cells that have the capability to direct the immune response of the host through the stimulation of B and T lymphocytes (4); probiotic strains that have the capability to affect the maturation of dendritic cells could therefore provide a potent modulation of the immune response. Christensen et al (12) investigated the ability of six different strains of lactobacilli to affect dendritic cell maturation by measuring cytokine production and surface marker expression. They were able to demonstrate that the irradiated bacteria had varying effects, with L. reuteri DSM 12246 exhibiting the weakest effect on most surface markers and cytokines measured. L. casei CHCC 3139, on the other hand, was shown to have the strongest effects on surface markers and IL- 23 12 of the strains measured. Interestingly, when L. reuteri was added along with L. casei, L. reuteri was able to inhibit the increase in pro-inflammatory cytokines lL-12, IL-6, TNF, and the surface marker CD86 observed with L. casei alone. Several other studies have also investigated the response of dendritic cells to certain strains of L. reuteri and found significant differences based on the strain tested. Mohamadzadeh et al (43) determined that L. reuteri ATCC 23272 was able to activate and induce maturation of dendritic cells and trigger an increase in pro-inflammatory lL-12 production, suggesting that the L. reuteri strain was shifting the dendritic cells towards polarization of a Th1 response. The authors observed similar effects with both live and irradiated cells, suggesting that bacterial viability was not essential for this effect. On the other hand, Smits et al (67) found no effect on cytokine production by dendritic cells when they were co- incubated with L. reuteri ASM 20016; however, they were able to determine that the dendritic cells had been “primed” to drive the development of T regulatory cells that then led to an increase in lL-10 production by T helper cells. lL-10 is typically considered to be involved in anti-inflammatory responses. The authors of this study also demonstrated differing effects based on the ratio of bacteria to dendritic cells. They showed that a ratio of bacteria to dendritic cells that was higher than 1:1 actually resulted in a decreased priming effect, suggesting that care should be taken when choosing the dosage amount of probiotics. These studies emphasize the need for thoroughly testing probiotic strains to determine their immunomodulatory effects before using them individually or in combination 24 as treatments, as some strains of L. reuteri promoted production of lL-12 (pro- inflammatory) and others promoted production of lL-10 (anti-inflammatory). Several labs have investigated the effect of probiotic administration on the production of the pro-inflammatory cytokine, tumor necrosis factor-alpha (TNF). Soria et al (68) showed that L. reuteri CRL 1098 was able to decrease TNF production, while L. acidophilus CRL 1014 and L. rhamnosus CRL 1036 increased production of TNF by human peripheral blood mononuclear cells, demonstrating again that some probiotic strains may be considered immunosuppressive, while others may be considered immunostimulatory. The effects on TNF production were only observed with live bacterial cells, and these effects were suggested to be somehow involved with lipid rafts. Lipid rafts are microdomains found in eukaryotic plasma membranes that have been shown to be involved in interactions with pathogenic bacteria. This study again suggests an overlap in the mechanisms through which probiotics and pathogens interact with eukaryotic cells, despite the fact that the outcomes of these interactions are Often very different. Although the previous study demonstrated a difference in the effects of probiotics of different species of Lactobacillus, variation in the effect of different strains of L. reuteri has also been observed. A study comparing three strains of L. reuten', ATCC PTA 6475, ATCC 55730, and CF48-3A, found that only L. reuteri ATCC PTA 6475 was able to suppress TNF production from LPS-activated monocytes 25 and macrophages (38). The suppression was shown to be due to secreted compounds produced by the bacteria, and the effect was demonstrated for both human cell lines and monocyte-derived macrophages isolated from Crohn's disease patients. Lin et al (38) offer evidence that suggests that the TNF suppression observed with cell-free supernatant from L. reuteri ATCC PTA 6475 may be caused by transcriptional regulation of TNF. The authors also note that there were differences in the responses of the isolated macrophages from different individuals, suggesting that the genetic background of the host may also play an important role in the interaction. A particularly intriguing observation is that cell-free conditioned medium from L. reuteri ATCC PTA 6475 was able to suppress TNF to similar levels as the current Crohn’s disease treatment, infiiximab (a TNF antibody), suggesting that probiotics may have potential as treatments for IBD (38). Animal studies and clinical trials. The most common studies performed on potential probiotic strains are in vitro tests to investigate whether the strains demonstrate the “necessary” characteristics to provide probiotic benefit, including survival to stresses in the gastrointestinal tract (acid and bile), the ability to attach to mucus or epithelial surfaces, the ability to inhibit or displace pathogens, and the ability to modify the host immune response (18, 54, 62). Although these characteristics are often considered to be important for a bacterial strain to be able to cause beneficial 26 effects, the mechanism of action for most of the beneficial effects attributed to probiotic strains is unknown, and therefore, what characteristics make a strain “effective” are also unknown. For example, Van Coillie et al (79) carefully screened strains of Lactobacillus isolated from hens for various characteristics such as production of antimicrobial acids and adherence to epithelial cells. Although they were able to identify a strain of L. reuteri that did decrease counts of Salmonella enterica from inoculated chicks, they reached the conclusion that the in vitro trait selection did not equal in vivo effectiveness. The authors suggest that this difference may be due to host-associated factors that are not included during in vitro testing (79). Therefore, the ultimate test for a potential probiotic strain comes in the form of in vivo studies, including animal models of disease, and ultimately, clinical trials. There are many animal studies and clinical trials involving L. reuteri; a selected few will be discussed here. Diarrhea. L. reuteri has been shown to be effective against diarrhea of various causes in children. Shornikova et al (65, 66) demonstrated that L. reuteri could actually be used as a treatment to reduce the duration of diarrhea (primarily from rotavirus) in children. In this study, the probiotic was administered after children were brought to the hospital for treatment for the diarrhea. In another study, it was found that children in a daycare that received L. reuteri ATCC 55730 had significantly less diarrhea episodes and the duration of these episodes was shorter than in the control group Of children (86). 27 Inflammatory bowel disease (IBD). Inflammatory bowel disease is a set of gastrointestinal disorders marked by chronic inflammation that affects a smaller portion of the population and is more severe than lBS. IBD can be divided into Crohn’s disease or ulcerative colitis, depending on the prevalence and location of inflammation. The prevalence of IBD has increased over the last few decades, particularly in regions with historically low occurrence (88). The proposed causes of IBD include: an overly aggressive immune response to commensal bacteria due to genetic factors or irregular immune response of the host, the presence or absence of particular bacteria, or environmental factors (58, 64). Probiotics, which are proposed to act through alteration of host immune responses or alteration of host microbial communities, are proposed as possible therapies for IBD (58, 64). Several studies have shown that lL-10 deficient mice, which are prone to spontaneous colitis unless raised in germ-free conditions, have a difference in their lactic acid bacteria population than wild-type control mice. Both Madsen (39) and Pena (48) found that the dominant Lactobacillus in wild-type control mice was L. reuteri, while the dominant type in lL-10 deficient mice was L. johnsonii. These studies not only suggest again that host genetic background can affect the microbial community, but also suggest that the presence of certain lactobacilli, including L. reuteri, may prevent spontaneous colitis. In fact, Madsen (39) was able to show that by repopulating lL-10 deficient mice with L. reuteri, the development of colitis was prevented, although the study also showed that 28 increasing the amount of lactic acid bacteria in the gut by lactulose treatment also prevented colitis, so the effect of L. reuteri therapy observed in that particular study may not be specific to L. reuteri. Probiotic L. reuteri has also been demonstrated to have some level of effectiveness against various rodent models of induced colitis, including Helicobacter hepaticus-induced (49), acetic acid-induced (28), and TNBS (trinitrobenzenesulfonic acid)-induced (50). Pena (49) showed that inflammation levels associated with H. hepaticus-induced colitis in lL-10 deficient mice were decreased by pre-treatment with L. reuteri 6798, a strain that had previously been shown to reduce TNF levels in vitro (48). Interestingly, the colonization levels of H. hepaticus were not affected, suggesting that immunomodulation may be the main mechanism of action observed in this study. These animal studies demonstrating a prevention or reduction of colitis with administration of L. reuteri indicates that certain strains may be able to prevent or alleviate colonic inflammation associated with inflammatory bowel disease (IBD). A one-month clinical trial investigated the effect of administration of a combination of L. reuteri RC-14 and L. rhamnosus GR-1 to individuals with IBD (either Crohn’s or ulcerative colitis) or healthy control subjects. Different effects of the probiotics were observed in both groups of individuals. One effect observed was an increase in CD4+ CD25high T cells in the IBD patients; this putatively immunosuppressive shift could help play a role in promoting and 29 maintaining remission in these patients (6). It is intriguing to note that probiotic effects may be altered in response to the health of the host. Hypercholesterolemia. Probiotics are also currently being investigated for the ability to lower serum cholesterol levels in the host. L. reuteri CRL 1098 has been shown to not only decrease cholesterol levels in mice, but also to prevent the development of hypercholesterolemia (72, 73). In the first study, hypercholesterolemic mice were fed L. reuteri CRL 1098 for seven days. After this treatment, a significant reduction in total cholesterol, along with a reduction in triglyceride levels and an increased ratio of HDL (high density lipoprotein) to LDL cholesterol was observed. The authors suggest that these observed effects may be due to either lower levels of intestinal absorption of cholesterol or higher catabolism of lipids (73). One other consideration would be the possibility that bile salt hydrolase activity of the bacteria resulted in the lower cholesterol levels (7), as deconjugated bile acids are more likely to be excreted, requiring the host to pull from serum cholesterol levels to synthesize new bile acids. Colic. L. reuteri ATCC 55730 has even been investigated as a probiotic for the alleviation of colic, an infantile disorder marked by excessive crying and irritability of unknown cause. When compared in a clinical trial to simethicone (a common colic treatment typically found to not be very effective), a reduction in crying time was reported for the probiotic-treated group over the 28-day course of the study. Crying time were reported at 51 minutes per day in the probiotic-treated group 30 and 145 minutes per day in the simethicone-treated group at the 28 day point, suggesting that probiotics may alleviate colicky symptoms (61 ). General health. Weizman et al (86) demonstrated that children in a daycare setting that received L. reuteri not only had significantly fewer episodes of fever and diarrhea than a control group, but also had significantly less clinical visits, daycare absences, and antibiotic prescriptions. In another study where L. reuteri ATCC 55730 was given to Swedish workers, improved health was found in the probiotic-treated group. The probiotic-treated group had less sick leave due to respiratory or gastrointestinal infections, and also had a lower frequency of sick days (measured as the number of sick days relative to the number of work days) than the control group (77). Oral health. L. reuteri has also been included in studies involving oral health, in part because of the potent antimicrobial activities of many of the strains. Many of the studies have involved determining the effects of probiotic administration on the levels of Streptococcus mutans, one of the main contributors to tooth decay. L. reuteri ATCC 55730 has been shown to have in vitro inhibitory activity against S. mutans; this antagonistic effect was also observed in human volunteers fed a yogurt product containing the strain. In these volunteers, a significantly lower population of S. mutans was isolated from the oral cavity, suggesting a decreased risk of caries (46). Caglar has also demonstrated the effectiveness of this strain by showing a significant reduction in salivary mutans streptococci 31 levels in volunteers that consumed L. reuteri ATCC 55370 in straw, tablet, or gum form (9, 10). Although the research in the probiotic field has increased exponentially over the past twenty years or so, there is still much that needs to be investigated. The research at this point includes many in vitro studies investigating survival of stresses that the bacteria will be exposed to in the gastrointestinal tract, the ability to inhibit pathogens, and the ability to modulate the host immune response. In vivo studies are investigating a wide range of disorders including bacterial infections, allergies, and gastrointestinal disorders. Although various strains of L. reuten' have demonstrated effectiveness in various in vitro and in vivo studies as discussed in this review, there are still large areas of research that need to be conducted. For example, there appears to be wide ranging differences between the physiology and effects of particular strains within the species, and very little has been determined about the actual interactions between L. reuteri and the host. This thesis focuses on characterization of different aspects Of L. reuteri physiology that may contribute to the probiotic potential of the species. Micorarray and mutational analysis of the bile stress response of L. reuteri ATCC 55730 identified several genes that play significant roles in either survival or growth of this strain in the presence of bile (Chapter 2). These genes may contribute to survival of the strain during passage through the host gastrointestinal tract. The study also identified a putative matrix metalloprotease 32 gene that is up-regulated in the presence of bile, but does not play a role in survival or growth of the strain in bile. The function of this gene is of particular interest, as it shows significant homology to eukaryotic matrix metalloproteases which are known to play important roles in the homeostasis of the gastrointestinal tract (Appendix C). The bile salt hydrolase activity in L. reuteri ATCC PTA 6475 was also investigated. The bacterial function Of this activity has yet to be elucidated, although the research presented in this thesis suggests that it may be involved in resistance to bile under acidic conditions (Chapter 3). An understanding of the role this activity plays in survival of the bacteria during passage through the gastrointestinal tract is important, as the activity has been proposed to also affect the host. The proposed effects on the host range from a (beneficial lowering of serum cholesterol levels to putatively harmful activation of carcinogens or malabsorption of fats and nutrients. Finally an investigation of the potential immunomodulatory role of lactobacillic acid, a cyclopropyl fatty acid that is specific to certain strains of L. reuteri at particular stages of growth is included. Certain strains of L. reuteri have been demonstrated to suppress TNF production from LPS-activated macrophages in vitro. This activity is proposed to be due to secreted molecules produced by these strains at early stationary to late stationary phases of growth. The production of lactobacillic acid is shown to be specific to strains with this 33 immunomodulatory activity, and the presence of this compound is shown to correlate with the growth stages during which the immunomodulatory compound is produced (Chapter 4). 34 10. REFERENCES Abachin, E., C. Poyart, E. Pellegrini, E. Milohanic, F. Fiedler, P. Berche, and P. Trieu-Cuot. 2002. Formation of D-alanyl-lipoteichoic acid is required for adhesion and virulence of Listeria monocytogenes. Mol. Microbiol. 43:1-14. Abbas Hilmi, H. T., A. Surakka, J. Apajalahti, and P. E. Saris. 2007. Identification of the most abundant Lactobacillus species in the crop of 1- and 5-week-old broiler chickens. Appl. Environ. Microbiol. 73:7867-7873. Axelsson, L. T., T. C. Chung, W. J. Dobrogosz, and S. E. Lindgren. 1989. Production of a broad spectrum antimicrobial substance by Lactobacillus reuteri. Microb. Ecol. Health Dis. 2:131-136. Banchereau, J., and R. M. Steinman. 1998. Dendritic cells and the control of immunity. Nature 392:245-252. Barbirato, F., J. P. Grivet, P. Soucaille, and A. Bories. 1996. 3- Hydroxypropionaldehyde, an inhibitory metabolite of glycerol fermentation to 1,3-propanediol by enterobacterial species. Appl. Environ. Microbiol. 62:1448-1451. Baroja, M. L., P. V. Kirjavainen, S. Hekmat, and G. Reid. 2007. Anti- inflammatory effects of probiotic yogurt in inflammatory bowel disease patients. Clin. Exp. Immunol. 149:470-479. Begley, M., C. Hill, and C. G. Gahan. 2006. Bile salt hydrolase activity in probiotics. Appl. Environ. Microbiol. 72:1729-1738. Cadieux, P., A. Wind, P. Sommer, L. Schaefer, K. Crowley, R. A. Britton, and G. Reid. 2008. Evaluation of reuterin production in urogenital probiotic Lactobacillus reuteri RC-14. Appl. Environ. Microbiol. 74:4645- 4649. Caglar, E., S. K. Cildir, S. Ergeneli, N. Sandalli, and S. Twetman. 2006. Salivary mutans streptococci and lactobacilli levels after ingestion of the probiotic bacterium Lactobacillus reuteri ATCC 55730 by straws or tablets. Acta. Odontol. Scand. 64:314-318. Caglar, E., S. C. Kavaloglu, O. O. Kuscu, N. Sandalli, P. L. Holgerson, and S. Twetman. 2007. Effect of chewing gums containing xylitol or probiotic bacteria on salivary mutans streptococci and lactobacilli. Clin. Oral Investig. 11:425-429. 35 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. Carey, C. M., M. Kostrzynska, S. tha, and S. Thompson. 2008. The effect of probiotics and organic acids on Shiga-toxin 2 gene expression in enterohemorrhagic Escherichia coli O157:H7. J. Microbiol. Methods 73:125-132. Christensen, H. R., H. Frokiar, and J. J. Pestka. 2002. Lactobacilli differentially modulate expression of cytokines and maturation surface markers in murine dendritic cells. J. Immunol. 168:171-178. Chung, T. C., L. T. Axelsson, S. E. Lindgren, and W. J. Dobrogosz. 1989. In vitro studies on reuterin synthesis by Lactobacillus reuteri. Microb. Ecol. Health Dis. 2:137-144. Cleusix, V., C. Lacroix, S. Vollenweider, M. Duboux, and G. Le Blay. 2007. Inhibitory activity spectrum of reuterin produced by Lactobacillus reuteri against intestinal bacteria. BMC Microbiol. 7:101. Collins, L. V., S. A. Kristian, C. Weidenmaier, M. Faigle, K. P. Van Kessel, J. A. Van Strijp, F. Gotz, B. Neumeister, and A. Peschel. 2002. Staphylococcus aureus strains lacking D-alanine modifications of teichoic acids are highly susceptible to human neutrophil killing and are virulence attenuated in mice. J. Infect. Dis. 186:214-219. Cotter, P. D., and C. Hill. 2003. Surviving the acid test: responses of gram-positive bacteria to low pH. Microbiol. Mol. Biol. Rev. 67:429-453. de Vrese, M., and J. Schrezenmeir. 2008. Probiotics, prebiotics, and synbiotics. Adv. Biochem. Eng. Biotechnol. 111:1-66. Dunne, C., L. Murphy, 8. Flynn, L. O'Mahony, S. O'Halloran, M. Feeney, D. Morrissey, G. Thornton, G. Fitzgerald, C. Daly, B. Kiely, E. M. Quigley, G. C. O'Sullivan, F. Shanahan, and J. K. Collins. 1999. Probiotics: from myth to reality. Demonstration of functionality in animal models of disease and in human clinical trials. Antonie Van Leeuwenhoek 76:279-292. FAO. 2001. Health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria: report of a joint FAONVHO expert consultation on evaluation of health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria. Food and Agriculture Organization. Forage, R. G., and E. C. Lin. 1982. DHA system mediating aerobic and anaerobic dissimilation of glycerol in Klebsiella pneumoniae NCIB 418. J. Bacteriol. 151 :591-599. 36 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. Forsberg, C. W. 1987. Production of 1,3-Propanediol from Glycerol by Clostridium acetobutylicum and Other Clostridium Species. Appl. Environ. Microbiol. 53:639-643. Géinzle, M. G., A. HOItzel, J. Walter, G. Jung, and W. P. Hammes. 2000. Characterization of reutericyclin produced by Lactobacillus reuteri LTH2584. Appl. Environ. Microbiol. 66:4325-4333. Ganzle, M. G., and R. F. Vogel. 2003. Contribution of reutericyclin production to the stable persistence of Lactobacillus reuteri in an industrial sourdough fermentation. Int. J. Food. Microbiol. 80:31-45. Ganzle, M. G., and R. F. Vogel. 2003. Studies On the mode of action of reutericyclin. Appl. Environ. Microbiol. 69: 1 305-1 307. Gunn, J. S. 2000. Mechanisms of bacterial resistance and response to bile. Microbes Infect. 2:907-13. Heinemann, C., J. E. van Hylckama Vlieg, D. B. Janssen, .H. J. Busscher, H. C. van der Mei, and G. Reid. 2000. Purification and characterization of a surface-binding protein from Lactobacillus fermentum RC-14 that inhibits adhesion of Enterococcus faecalis 1131. FEMS Microbiol. Lett. 190:177-180. Hofman, A. 1998. Bile secretion and the enterohepatic circulation of bile acids, p. 937-948. In M. Feldman, B. Scharschmidt, and M. Sleisenger (ed.), Sleisengers and Fordtran's Gastrointestinal and Liver Disease, 6 ed. W. B. Saunders, Co., Philadelphia. Holma, R., P. Salmenpera, J. Lohi, H. Vapaatalo, and R. Korpela. 2001. Effects of Lactobacillus rhamnosus GG and Lactobacillus reuteri R2LC on acetic acid-induced colitis in rats. Scand. J. Gastroenterol. 6:630-635. H6ltzel, A., M. G. Ganzle, G. J. Nicholson, W. P. Hammes. and G. Jung. 2000. The first low molecular weight antibiotic from lactic acid bacteria: reutericyclin, a new tetramic acid. Angew. Chem. Int. Ed. 39:2766-2768. Jeléié, l., E. Htifner, H. Schmidt, and C. Hertel. 2008. Repression of the locus of the enterocyte effacement-encoded regulator of gene transcription of Escherichia coli O157:H7 by Lactobacillus reuteri culture supernatants is LuxS and strain dependent. Appl. Environ. Microbiol. 74:3310-3314. 37 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. Jonsson, H., E. StrOm, and S. Roos. 2001. Addition of mucin to the growth medium triggers mucus-binding activity in different strains of Lactobacillus reuteri in vitro. FEMS Microbiol. Lett. 204:19-22. Kabuki, T., T. Saito, Y. Kawai, J. Uemura, and T. ltoh. 1997. Production, purification and characterization of reutericin 6, a bacteriocin with lytic activity produced by Lactobacillus reuteri LA6. Int. J. Food. Microbiol. 34:145-156. Kawai, Y., Y. lshii, K. Arakawa, K. Uemura, B. Saitoh, J. Nishimura, H. Kitazawa, Y. Yamazaki, Y. Tateno, T. Itoh, and T. Saito. 2004. Structural and functional differences in two cyclic bacteriocins with the same sequences produced by lactobacilli. Appl. Environ. Microbiol. 70:2906-291 1. Klaenhammer, T. R. 1982. Microbiological considerations in selection and preparation of Lactobacillus strains for use as dietary adjuncts. J. Dairy Sci. 65: 1 339-1 349. Larsen, N., P. Nissen, and W. G. Willats. 2007. The effect of calcium ions on adhesion and competitive exclusion of Lactobacillus ssp. and E. coli 0138. Int. J. Food Microbiol. 114:113-119. Laughton, J. M., E. Devillard, D. E. Heinriches, G. Reid, and J. K. McCormick. 2006. Inhibition of expression of a staphylococcal superantigen-like protein by a soluble factor from Lactobacillus reuteri. Microbiology 1 52:1155-1167. Lee, K., H. G. Lee, K. Pi, and Y. J. Choi. 2008. The effect of low pH on protein expression by the probiotic bacterium Lactobacillus reuteri. Proteomics 8:1624-1630. Lin, Y. P., C. H. Thibodeaux, J. A. Pefia, G. D. Ferry, and J. Versalovic. 2008. Probiotic Lactobacillus reuteri suppress proinflammatory cytokines via c-Jun. lnflamm. Bowel Dis. 14:1068-1083. Madsen, K. L., J. S. Doyle, L. D. Jewell, M. M. Tavernini, and R. N. Fedorak. 1999. Lactobacillus species prevents colitis in interleukin 10 gene-deficient mice. Gastroenterology 116:1 107-1 1 14. Martin, R., M. Olivares, M. L. Marin, J. Xaus, L. Fernandez, and J. M. Rodriguez. 2005. Characterization of a reuterin-producing Lactobacillus coryniformis strain isolated from a goat's milk cheese. Int. J. Food. Microbiol. 104:267-277. 38 41. 42. 43. 44. 45. 46. 47. 48. 49. McCoy, S., and S. E. Gilliland. 2007. Isolation and characterization of Lactobacillus species having potential for use as probiotic cultures for dogs. J. Food Sci. 72:M94-M97. Miyoshi, Y., S. Okada, T. Uchimura, and E. Satoh. 2006. A mucus adhesion promoting protein, MapA, mediates the adhesion of Lactobacillus reuteri to Caco-2 human intestinal epithelial cells. Biosci. Biotechnol. Biochem. 70: 1 622-1 628. Mohamadzadeh, M., S. Olson, W. V. Kalina, G. Ruthel, G. L. Demmin, K. L. Warfield, S. Bavari, and T. R. Klaenhammer. 2005. Lactobacilli activate human dendritic cells that skew T cells toward T helper 1 polarization. Proc. Natl. Acad. Sci. U S A 102:2880-2885. Morita, H., H. Toh, S. Fukuda, H. Horikawa, K. Oshima, T. Suzuki, M. Murakami, S. Hisamatsu, Y. Kato, T. Takizawa, H. Fukuoka, T. Yoshimura, K. ltoh, D. J. O'Sullivan, L. L. McKay, H. Ohno, J. Kikuchi, T. Masaoka, and M. Hattori. 2008. Comparative genome analysis of Lactobacillus reuteri and Lactobacillus fermentum reveal a genomic island for reuterin and cobalamin production. DNA Res. 15:151-161. Mukai, T., T. Asasaka, E. Sato, K. Mori, M. Matsumoto, and H. Ohori. 2002. Inhibition of binding of Helicobacter pylori to the glycolipid receptors by probiotic Lactobacillus reuteri. FEMS Immunol. Med. Microbiol. 32:105- 1 10. Nikawa, H., S. Makihira, H. Fukushima, H. Nishimura, Y. Ozaki, K. lshida, S. Darmawan, T. Hamada, K. Hara, A. Matsumoto, T. Takemoto, and R. Aimi. 2004. Lactobacillus reuteri in bovine milk fermented decreases the oral carriage of mutans streptococci. Int. J. Food. Microbiol. 95:219-223. Northfield, T. C., and I. McColl. 1973. Postprandial concentrations of free and conjugated bile acids down the length of the normal human small intestine. Gut 14:513-518. Pefia, J. A., S. Y. Li, P. H. Wilson, S. A. Thibodeau, A. J. Szary, and J. Versalovic. 2004. Genotypic and phenotypic studies of murine intestinal lactobacilli: species differences in mice with and without colitis. Appl. Environ. Microbiol. 70:558-568. Pefia, J. A., A. B. Rogers, Z. Ge, V. Ng, S. Y. Li, J. G. Fox, and J. Versalovic. 2005. Probiotic Lactobacillus spp. diminish Helicobacter hepaticus-induced inflammatory bowel disease in interleukin-10-deficient mice. Infect. Immun. 73:912-920. 39 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. Peran, L., S. Sierra, M. Comalada, F. Lara-Villoslada, E. Bailén, A. Nieto, A. Concha, M. Olivares, A. Zarzuelo, J. Xaus, and J. Galvez. 2007. A comparative study of the preventative effects exerted by two probiotics, Lactobacillus reuteri and Lactobacillus fermentum, in the trinitrobenzenesulfonic acid model of rat colitis. Br. J. Nutr. 97:96-103. Poyart, C., E. Pellegrini, M. Marceau, M. Baptista, F. Jaubert, M. C. Lamy, and P. Trieu-Cuot. 2003. Attenuated virulence of Streptococcus agalactiae deficient in D-alanyl-lipoteichoic acid is due to an increased susceptibility to defensins and phagocytic cells. Mol. Microbiol. 49:1615- 1625. Reuter, G. 2001. The Lactobacillus and Bifidobacterium microflora of the human intestine: composition and succession. Curr. Issues lntest. Microbiol. 2:43-53. Riley, M. A., and J. E. Wertz. 2002. Bacteriocins: evolution, ecology, and application. Annu. Rev. Microbiol. 56:117-137. Rolfe, R. D. 2000. The role of probiotic cultures in the control of gastrointestinal health. J. Nutr. 130:3968—4028. Roos, 8., P. Aleljung, N. Robert, B. Lee, T. Wadstrém, M. Lindberg, and H. Jonsson. 1996. A collagen binding protein from Lactobacillus reuteri is part of an ABC transporter system? FEMS Microbiol. Lett. 144:33-38. Roos, 8., and H. Jonsson. 2002. A high-molecular—mass cell-surface protein from Lactobacillus reuteri 1063 adheres to mucus components. Microbiology 148:433-442. Ryan, K. A., T. Jayaraman, P. Daly, C. Canchaya, S. Curran, F. Fang, E. M. Quigley, and P. W. O'Toole. 2008. Isolation of lactobacilli with probiotic properties from the human stomach. Lett. Appl. Microbiol. 47:269-274. Sartor, R. B. 2004. Therapeutic manipulation of the enteric microflora in inflammatory bowel diseases: antibiotics, probiotics, and prebiotics. Gastroenterology 126:1620-1633. Saunders, S., A. Bocking, J. Challis, and G. Reid. 2007. Effect of Lactobacillus challenge on Gardnerella vagina/is biofilms. Colloids Surf. B Biointerfaces 55:138-142. Sauvageot, N., K. Gouffi, J. M. Laplace, and Y. Auffray. 2000. Glycerol metabolism in Lactobacillus collinoides: production of 3- 40 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. hydroxypropionaldehyde, a precursor of acrolein. Int. J. Food. Microbiol. 55:167-170. Savino, F., E. Pelle, E. Palumeri, R. Oggero, and R. Miniero. 2007. Lactobacillus reuteri (American Type Culture Collection Strain 55730) versus simethicone in the treatment of infantile colic: a prospective randomized study. Pediatrics 119:e124-e130. Saxelin, M., S. Tynkkynen, T. Mattila-Sandholm, and W. M. de V05. 2005. Probiotic and other functional microbes: from markets to mechanisms. Curr. Opin. Biotechnol. 16:204-11. Schaefer, L., T. Auchtung, and R. Britton. The antimicrobial compound reuterin inhibits thiol groups and induces oxidative stress. Unpublished. Shell, 8., F. Shanahan, and L. O'Mahony. 2007. Probiotic effects on inflammatory bowel disease. J. Nutr.:8198—824S. Shornikova, A. V., I. A. Casas, E. lsolauri, H. Mykkanen, and T. Vesikari. 1997. Lactobacillus reuteri as a therapeutic agent in acute diarrhea in young children. J. Pediatr. Gastroenterol. Nutr. 24:399-404. Shornikova, A. V., I. A. Casas, H. Mykkanen, E. Salo, and T. Vesikari. 1997. Bacteriotherapy with Lactobacillus reuteri in rotavirus gastroenteritis. Pediatr. Infect. Dis. J. 16:1103-1 107. Smits, H. H., A. Engering, D. van der Kleij, E. C. de Jong, K. Schipper, T. M. van Capel, B. A. Zaat, M. Yazdanbakhsh, E. A. Wierenga, Y. van Kooyk, and M. L. Kapsenberg. 2005. Selective probiotic bacteria induce lL-10-producing regulatory T cells in vitro by modulating dendritic cell function through dendritic cell-specific intercellular adhesion molecule 3- grabbing nonintegrin. J. Allergy Clin. Immunol. 115:1260-1267. Soria, M., M. Mechoud, D. E. Q. A. Martin, D. E. V. G. Font, and V. R. A. 2008. Disruption of lipid rafts enhances the effect of lactobacilli on the production of tumor necrosis factor-alpha in mononuclear blood cells. Biosci. Biotechnol. Biochem. 72:3293-3296. Spinler, J. K., M. Taweechotipatr, C. L. Rognerud, C. N. 0u, S. Tumwasorn, and J. Versalovic. 2008. Human-derived probiotic Lactobacillus reuteri demonstrate antimicrobial activities targeting diverse enteric bacterial pathogens. Anaerobe 14: 1 66-1 71. Talarico, T. L., and W. J. Dobrogosz. 1989. Chemical characterization of an antimicrobial substance produced by Lactobacillus reuteri. Antimicrob. Agents Chemother. 33:674-679. 41 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. Taranto, M. P., M. L. Fernandez Murga, G. Lorca, and G. F. de Valdez. 2003. Bile salts and cholesterol induce changes in the lipid cell membrane of Lactobacillus reuteri. J. Appl. Microbiol. 95:86-91. Taranto, M. P., M. Medici, G. Perdigon, A. P. Ruiz Holgado, and G. F. Valdez. 2000. Effect of Lactobacillus reuteri on the prevention of hypercholesterolemia in mice. J. Dairy Sci 83:401-403. Taranto, M. P., M. Medici, G. Perdigon, A. P. Ruiz Holgado, and G. F. Valdez. 1998. Evidence for hypocholesterolemic effect of Lactobacillus reuteri in hypercholesterolemic mice. J. Dairy Sci. 81 :2336-2340. Taranto, M. P., F. Sesma, and G. F. de Valdez. 1999. Localization and primary characterization of bile salt hydrolase from Lactobacillus reuteri. Biotechnol. Lett. 21:935-938. Toba, T., S. K. Samant, E. Yoshioka, and T. ltoh. 1991. Reutericin 6, a new bacteriocin produced by Lactobacillus reuteri LA 6. Lett. Appl. Microbiol. 13:281-286. Todoriki, K., T. Mukai, S. Sato, and T. Toba. 2001. Inhibition of adhesion of food—borne pathogens to Caco-2 cells by Lactobacillus strains. J. Appl. Microbiol. 91 :154-159. Tubelius, P., V. Stan, and A. Zachrisson. 2005. Increasing work-place healthiness with the probiotic Lactobacillus reuteri: a randomised, double- blind placebo-controlled study. Environ. Health 4:25. Valeur, N., P. Engel, N. Carbajal, E. Connolly, and K. Ladefoged. 2004. Colonization and immunomodulation by Lactobacillus reuteri ATCC 55730 in the human gastrointestinal tract. Appl. Environ. Microbiol. 70:1176- 1 181. Van Coillie, E., J. Goris, l. Cleenwerck, K. Grijspeerdt, N. Botteldoorn, F. Van lmmerseel, J. De Buck, M. Vancanneyt, J. Swings, L. Herman, and M. Heyndrickx. 2007. Identification of lactobacilli isolated from the cloaca and vagina of laying hens and characterization for potential use as probiotics to control Salmonella Enteritidis. J. Appl. Microbiol. 102:1095- 1 106. Vesterlund, S., M. Karp, S. Salminen, and A. C. Ouwehand. 2006. Staphylococcus aureus adheres to human intestinal mucus but can be displaced by certain lactic acid bacteria. Microbiology 152:1819-1826. 42 81. 82. 83. 84. 85. 86. 87. 88. 89. Wall, T., K. Bath, R. A. Britton, H. Jonsson, J. Versalovic, and S. Roos. 2007. The early response to acid shock in Lactobacillus reuteri involves the CIpL chaperone and a putative cell wall-altering esterase. Appl. Environ. Microbiol. 73:3924-3935. Walter, J., P. Chagnaud, G. W. Tannock, D. M. Loach, F. Dal Bello, H. F. Jenkinson, W. P. Hammes. and C. Hertel. 2005. A high-molecular- mass surface protein (Lsp) and methionine sulfoxide reductase B (MsrB) contribute to the ecological performance of Lactobacillus reuteri in the murine gut. Appl. Environ. Microbiol. 71:979-986. Walter, J., D. M. Loach, M. Alqumber, C. Rockel, C. Hermann, M. Pfitzenmaier, and G. W. Tannock. 2007. D-alanyl ester depletion of teichoic acids in Lactobacillus reuteri 100-23 results in impaired colonization of the mouse gastrointestinal tract. Environ. Microbiol. 9:1750-1760. Walter, J., C. Schwab, D. M. Loach, M. G. Ganzle, and G. W. Tannock. 2008. Glucosyltransferase A (thA) and inulosucrase (lnu) of Lactobacillus reuteriTMW1.106 contribute to cell aggregation, in vitro biofilm formation, and colonization of the mouse gastrointestinal tract. Microbiology 154:72- 80. Wang, 8., H. Wei, J. Yuan, Q. Li, Y. Li, N. Li, and J. Li. 2008. Identification of a surface protein from Lactobacillus reuten' JCM1081 that adheres to porcine gastric mucin and human enterocyte-like HT-29 cells. Curr. Microbiol. 57:33-38. Weizman, Z., G. Asli, and A. Alsheikh. 2005. Effect of a probiotic infant formula on infections in child care centers: comparison of two probiotic agents. Pediatrics 115:5—9. Whitehead, K., J. Versalovic, S. Roos, and R. A. Britton. 2008. Genomic and genetic characterization of the bile stress response of probiotic Lactobacillus reuteri ATCC 55730. Appl. Environ. Microbiol. 74:1812-1819. Xavier, R. J., and D. K. Podolsky. 2007. Unravelling the pathogenesis of inflammatory bowel disease. Nature 448:427-434. Yun, J. H., K. B. Lee, Y. K. Sung, E. B. Kim, H. G. Lee, and Y. J. Choi. 2008. Isolation and characterization of potential probiotic lactobacilli from pig feces. J. Basic Microbiol. 48:1-7. 43 CHAPTER 2 GENOMIC AND GENETIC CHARACTERIZATION OF THE BILE STRESS RESPONSE OF PROBIOTIC LACTOBACILLUS REUTERI ATCC 55730 These results have been published in the article: Whitehead, K., J. Versalovic, S. Roos, and RA. Britton. 2008. Genomic and genetic characterization of the bile stress response of probiotic Lactobacillus reuteri ATCC 55730. Appl. Environ. Microbiol. Vol. 74, pgs 1812-1819. INTRODUCTION The idea that bacteria could benefit human health was postulated almost one hundred years ago by Elie Metchnikoff (23). Recently the use of probiotics, which are defined as “live microorganisms which when administered in adequate amounts confer a health benefit on the host” (10), has become increasingly popular. Probiotic microorganisms are currently being investigated for many possible health benefits in many different ailments including inflammatory bowel disease, diarrhea and hypercholesterolemia (9, 33, 40, 46). The mechanisms through which probiotics confer their beneficial effects are mostly unknown; examples of current theories include immunomodulation of the host by the synthesis of immunomodulatory compounds, the production of antimicrobial 44 compounds that inhibit pathogen growth, and large-scale alterations of the microbiota (28, 31). Probiotic bacteria encounter a variety of stresses that need to be overcome to remain viable. For example, many bacteria are packaged into food products such as yogurt and fermented milk, which exposes them to temperature and osmotic stress. After ingestion, probiotics must be able to survive the extreme acidic conditions in the stomach and the detergent properties of bile acids in the small intestine. Bile acids are amphipathic molecules that are synthesized from cholesterol and play an important role in the digestion of fats and absorption of fat-soluble vitamins. The concentration of bile acids ranges from 0.2 to 2% in the human small intestine and fluctuates based on the amount of fat intake in the diet (14). Bile acids have potent antimicrobial activity against many microbes and are known to cause damage to cells that are considered to be bile resistant, most likely via disruption of the membrane and cell wall. The resistance mechanisms of gram-negative bacteria are fairly well-characterized; these mechanisms include protection by the hydrophobic outer membrane and utilization of efflux pumps to expel bile salts that do enter the cell (13). The resistance mechanisms of gram-positive organisms, which in general are less bile-resistant than gram- negative bacteria, are less well understood. Lactic acid bacteria, particularly lactobacilli, are the genus most commonly used as probiotics, in part because of their safe usage in food production. Potential 45 new probiotic strains should include several important characteristics; they should be of human origin, non-pathogenic, able to remain viable in the gastrointestinal tract for at least short periods of time, and be resistant to various stresses (9). In the GI tract the main sources of antimicrobial stress are the low pH encountered in the stomach and the detergent like properties of bile acids found in the small intestine. Although the precise mechanisms by which bile acids cause cell death are not understood, their chemical nature indicates they will be able to solubilize membranes and cause significant membrane damage. This is supported by genetic and genomic studies in a variety of different species that show the main response of gram-positive organisms to bile exposure appears to be alteration of the cellular envelope. Isolation of bile-sensitive mutants of Enterococcus faecalis and Listeria monocytogenes identified genes mainly involved in maintenance and synthesis of the cell membrane and wall, as well as genes involved in general stress responses (2, 16). Microarray analysis of Lactobacillus plantarum and Lactobacillus acidophilus identified expression changes in genes whose product is found in the cell envelope (6, 25). In addition there is also microscopic evidence supporting the role of bile in alteration of the cellular envelope. Bron et al. demonstrated that cultures of L. plantarum cells exposed to bile contained some shrunken cells and cells that tended to clump together and had rough surfaces (5), while bile exposure also caused the appearance of shrunken and empty cells in cultures of Propionibacterium freudenreichii (1 7). 46 Lactobacillus reuteri is a species with a broad host range, with isolates originating from many different species including humans, pigs, chickens, dogs, mice, and hamsters (7). L. reuteri is also considered to be indigenous to the human GI tract (27). L. reuteri ATCC 55730, a strain currently marketed for probiotic usage, has been demonstrated in clinical trials to be effective against diarrhea in children, as well as to alleviate colic in infants (26, 30, 45). In addition, consumption of L. reuteri ATCC 55730 reduced the number of sick days taken by workers in a large trial in Sweden (38). How these benefits are achieved at the molecular level is still unknown. L. reuteri ATCC 55730 is known to produce a broad-spectrum antimicrobial compound, reuterin, by metabolism of glycerol under anaerobic conditions (36). Based on the observation that this strain is able to survive in the human duodenum and ileum (41), it is an appropriate organism to use in the investigation of bile resistance mechanisms. This research investigated the gene expression response of L. reuteri ATCC 55730 to bile exposure and has begun to uncover the mechanisms this strain uses to survive and grow in the presence of bile. Microarray experiments were conducted to determine the gene expression profiles of cells upon initial bile exposure (bile shock) and cells that had resumed growth in the presence of bile (bile adaptation). Based on the microarray results, nine genes were chosen for mutational analysis. These results indicate some of the mechanisms important in the bile shock and adaptation responses of L. reuteri ATCC 55730 in vitro and 47 may provide a further understanding of characteristics important for survival in the gastrointestinal tract. MATERIALS AND METHODS Bacterial strains and growth conditions. The bacterial strains and plasmids used in this study are listed in Table 2.1. All liquid cultures of lactobacilli were grown under microaerobic conditions (2% 02, 5% C02, balanced with N2) in MRS broth (BD Difco) at 37°C, unless othen~ise specified. All plate cultures of lactobacilli were grown under anaerobic conditions using the GasPack EZ Anaerobe Container system (BD Difco) at 37°C, unless othenrvise specified. Lactobacillus reuteri strains containing pVE6007 were grown at 35°C. All E. coli was grown under aerobic conditions at 37°C in LB broth (BD Difco). When specified, drugs were added to the following concentrations: 10 ug/ml (L. reuteri) or 400 ug/ml (E. coli) erythromycin, 10 pig/ml chloramphenicol, and 40 pg/ml kanamycin. L. reuteri mutants (containing the p0Rl28 disruption) were always grown in the presence of 10 ug/ml erythromycin. Dehydrated bovine bile/ox gall (Sigma) was resuspended in MRS broth to make a 50% weight/volume solution. This mixture was sterilized by autoclaving and stored at 37°C for up to four weeks. 48 Table 2.1. Bacterial strains and plasmids used for this study. Strain or Reference plasmid Description or source Strains E. coli strain containing a chromosomal copy E01000 of the pWV01 repA gene; Kanr (15) L. reuteri strain isolated from human breast Biogaia, AB, ATCC 55730 milk Sweden L. reuteri ATCC 55730 clpL (lr1864) mutant, PRB190 Emr (42) L. reuteri ATCC 55730 putative esterase PRB188 (lr1516) mutant, Emr (42) lr0085::pKW01 in an ATCC 55730 PRB167 background, Emr This study L. reuteri ATCC 55730 clpE (lr0004) mutant, PRB186 Emr Stefan Roos lr1265::pKW02 in an ATCC 55730 PRB126 background, Emr This study lr1584::pKW03 in an ATCC 55730 PRB130 background, Emr This study lr1291::pKW04 in an ATCC 55730 PRB163 background, Emr This study lr1351::pKW05 in an ATCC 55730 PRB125 background, Emr This study lr1706::pKW06 in an ATCC 55730 PRB114 background, Emr This study Plasmids Cmr repA-positive temperature-sensitive pVE6007 derivative of pWV01 (19) p0Rl28 Emr repA-negative derivative of pWV01 (15) pKW01 p0Rl28 + 203 bp insert from lr0085 This study pKW02 ORI28 + 303 bp insert from Ir1265 This study KW03 gORI28 + 331 bp insert from lr1584 This study pKW04 p0Rl28 + 310 bp insert from lr1291 This study pKW05 p0Rl28 + 152 bp insert from lr1351 This study pKW06 p0Rl28 + 240 bp insert from lr1706 This study 49 RNA isolation. For each of five biological replicate experiments, a culture of L. reuteri ATCC 55730 was grown in MRS broth to an optical density at 600 nm (0.0.600) approximately equal to 0.5. Upon reaching this stage in growth, 0.5% oxgall was added to the culture. At the correct time points, 5 ml samples were collected from the culture and immediately mixed with an equal part of ice-cold methanol. Cell pellets were collected by centrifugation, washed with STE buffer (6.7% sucrose; 50 mM Tris-Cl, pH 8.0; 1 mM EDTA), and resuspended in STE buffer containing 0.25 units/pl mutanolysin (Sigma). Cells pellets were then incubated at 37°C for 20 minutes. RNA was then isolated using the Qiagen RNeasy Kit according to manufacturer’s instructions. Microarray experiments. Long oligonucleotides (60-mers) were designed and synthesized for 1864 open reading frames from a draft genome sequence of L. reuteri ATCC 55730 (1) and 15 open reading frames encoding known extracellular proteins from L. reuteri DSM 20016 (43) using OligoArray 1.0 Software. Six control 60-mer oligonucleotides were also included. These controls are identical to DNA sequences from E. coli genes (yacF, ybaS, yciC, yfiF, yng, and ych) and have no sequence similarity to the L. reuteri genome. Once synthesized the oligonucleotide concentrations were normalized to a concentration of 25 uM and spotted onto Corning UltraGAPS-ll slides using an OmniGrid Robot (GeneMachines). Each gene was represented once on the microarray. All six of the control spots were represented 8 times on the array, once in each subgrid. Oligonucleotide design, synthesis, and array construction 50 were performed at the Research Technology Support Facility at Michigan State University, East Lansing, MI, USA. RNA isolation, labeling, and hybridization were carried out essentially as previously described (39, 42). (Information regarding the microarray platform can be found at NCBls Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.g)v/geo/) under GEO platform number GPL6366). Five biological replicates were performed for each of the two sets of microarray experiments. In addition, technical replication was achieved by switching the dyes used for labeling each biological replicate. Therefore, each RNA sample was subjected to two hybridizations and values used for subsequent data analysis were averages of the dye swap values. The first set of experiments, referred to as the bile shock experiments, compared the gene expression profiles of cells before exposure to 0.5% bile to those that had been exposed for 15 minutes. The second set of experiments, referred to as the bile adaptation experiments, compared the gene expression profiles of cells before exposure to 0.5% bile to those that had begun growing again after exposure (Figure 2.1). 51 0.1 - 0.0.600 0.01 I I I T ‘T I I 0 100 200 300 400 500 600 700 time (min) Figure 2.1. Representative growth curve of L. reuteri ATCC 55730 used for microarray experiments (arrows represent time points where samples were taken for RNA isolation). Open arrows represent samples for bile shock experiments; filled arrows represent samples for bile adaptation experiments. 0.5% oxgall was added at 250 minutes. Microarray data was analyzed using iterative outlier analysis with three iterations as previously described (4, 39). Briefly, iterative outlier analysis calculates the geometric mean and standard deviation of the entire dataset. Differentially expressed genes (outliers) were selected as being more than 2.5 standard deviations away from the mean of the population. To identify additional differentially expressed genes in the dataset. the outliers were removed and the geometric mean and standard deviations were recalculated and any genes that 52 were more than 2.5 standard deviations from the mean were identified as differentially expressed. Mutant construction. Mutants were created using the system developed by Russell and Klaenhammer (29) and modified for use in L. reuteri by Walter et al (44). Briefly, 200 - 300 bp from the gene of interest were PCR-amplified from L. reuteri ATCC 55730 and cloned into pORl28. The plasmid with the insertion was then transformed into E. coli EC1000, a carrier strain that contains the RepA protein needed for pORl28 to replicate; the transformed cells were grown in the presence of erythromycin and kanamycin (EC1000). pORl28 with the insertion was then extracted and transformed into L. reuteri ATCC 55730 cells containing pVE6007. pVE6007 is a helper plasmid that provides RepA (also allowing p0Rl28 to replicate). L. reuteri cells containing both plasmids were grown aerobically without shaking at the permissive temperature of 35°C in the presence of chloramphenicol and erythromycin for 18 hours. This culture was then diluted 1:200 and grown aerobically in the presence of erythromycin without shaking at 45°C for 8-24 hours. The 45°C culture was then plated onto MRS + erythromycin plates and incubated at 45°C for 24 hours. Isolated colonies were then obtained by streaking onto fresh IVIRS + erythromycin plates and incubated at 45°C for another 24 hours to ensure loss of pVE6007. Individual colonies were then selected for integration of pORl28 based on erythromycin resistance and screened for chloramphenicol sensitivity to confirm loss of pVE6007 at 37°C. Colonies that had lost pVE6007 were then screened for the correct insertion by 53 PCR-amplification of both flanking regions (one primer annealing to the chromosome outside of the region cloned into pORl28 and one primer annealing to pORl28) and confirmation of the absence of the correctly sized wild-type gene (also through PCR). PCR primers are available by request. Bile stress assays. To determine levels of bile resistance for the mutants, the percent survival of wild-type and mutant cultures was determined after 30 minutes of exposure to 0.3% bile. In short, cultures were grown under microaerobic conditions in MRS at 37°C to an O.D.500 = 0.5. Samples were taken and colony counts were determined by dilution plating. 0.3% oxgall was then added to each culture, and after 30 minutes, colony counts were again determined. The before bile and after bile colony counts were used to determine percent survival for each strain. All growth curves and viability plating were performed three times for each strain, with the exception of the wild-type experiments, which were repeated eight times. RESULTS Lactobacillus reuteri ATCC 55730 is able to grow in physiologically relevant concentrations of bile. One important characteristic for probiotic bacterial strains is the ability to remain viable during passage through the gastrointestinal tract, including the ability to overcome exposure to bile stress in the small intestine. Growth experiments were conducted to determine the response of L. 54 reuteri ATCC 55730 to physiological concentrations of bovine bile. In general, when 0.05 to 0.1% bile was added to an early or mid-log culture (0.0600 = 0.2 or 0.5), the culture continued growing, although at a slightly reduced rate. ' The doubling time of the culture would slow from 38 minutes before the addition of bile to 50 minutes after the addition of bile. When concentrations of bile ranging from 0.3% to 5% were added, we observed a period of growth arrest followed by a resumption of growth. However the doubling time in the presence of these higher bile concentrations was 3-4 times slower than prior to treatment with bile. The growth-phase of L. reuteri cells also influenced the ability of bile to affect cell growth and viability. Early log-phase cells (ODsoo = 0.2) were the most resistant to the effects of bile treatment. Addition of bile at later stages of log-phase growth and early stationary phase indicated that cells (become more susceptible to bile as the culture density increases (as measured by a decrease in the optical density and cell viability of the culture after the addition of bile). Active growth appears to be required for this effect as late stationary phase cultures were completely resistant to bile stress, even at concentrations of 5%. Microarray analysis of genes involved in bile shock and adaptation. We used DNA microarrays to characterize both the bile shock response and bile adaptation response of L. reuteri. When cells encounter stress they often respond by altering their gene expression program to effectively counteract stress-induced damage. Because L. reuteri exhibits a biphasic response to bile 55 exposure we measured the global RNA profiles of cells that were paused for growth (which we denote as bile shock) and cells that had resumed growth in the presence of bile (bile adaptation). i. Bile shock. In order to determine the genes involved in the bile shock response, microarray experiments were carried out to compare the gene expression profiles of mid-log cells that had not been exposed to bile to cells that had been exposed to 0.5% bile for 15 minutes. Eighty-eight genes were found to have significant expression changes, with 45 genes over-expressed and 43 genes under-expressed after 15 minutes of bile exposure. The majority of under- expressed genes are classified as being involved in substrate transport and metabolism, which is expected due to the lack of growth observed upon exposure to bile (Table 2.2). The over-expressed genes are found in a wide variety of classes and several have known roles in adaptation to other types of stresses (see below and discussion). 56 Table 2.2. Classesa of genes differentially expressed during the first 15 minutes of exposure to 0.5% bile. Number of genes Number of genes over-expressed under-expressed Gene classification during bile shock duringbile shock Energy production and conversion 0 4 Cell division and . . b envelope biogeneSIs 1 3 Substrate transport and metabolismC 4 21 Translation, ribosomal structure and biogenesis 2 2 Transcription 8 1 Replication, recombination and repair 3 3 Posttranslational modification, protein turnover, chaperones 6 2 Defense mechanisms 3 0 Unknown functions 18 7 Total 45 43 aGenes were classified based on COG domains found in the protein sequence through a search of the JGI Integrated Microbial Genomes database. b . . . . . This class represents two COG categories: cell cycle control, cell lelSlOl‘I, and chromosome partitioning and cell wall/membrane/envelope biogenesis. CThis class represents multiple COG categories that include transport and metabolism of carbohydrates, amino acids, nucleotides, coenzymes, lipids, inorganic ions, and secondary metabolites. ii. Bile adaptation. The expression profiles of mid-log cells that had not been exposed to bile were also compared to the profiles of cells that had resumed growth in the presence of 0.5% bile. After analysis of this set of array experiments, 84 genes were found to have significant expression changes, with 57 17 being over-expressed during the adaptation stage and 67 being under- expressed. Again, the majority of under-expressed genes during growth in bile are classified as being involved in substrate transport and metabolism; in addition, genes involved in energy production, translation and ribosome structure and biogenesis, and genes of unknown function are also under-expressed. Most of these changes are likely due to the dramatic reduction in growth rate of L. reuteri cells grown in the presence of bile. There were 17 genes that were significantly over-expressed during growth in bile; over half of these genes are annotated as having unknown functions (Table 2.3). The expression patterns of a subset of genes that were differentially expressed were found to overlap between the two sets of microarray experiments, with eight genes being over- expressed and 12 genes being under-expressed during both bile shock and adaptation. (A full description of the differentially expressed genes discovered in all of the microarray experiments can be found in Supplementary Tables 2.7 to 2.10 (located at the end of this chapter). A complete dataset of microarray data can be found at NCBls Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/qeol) under GEO Series accession number GSE10155). 58 Table 2.3. Classesa of genes differentially expressed during growth in the presence of 0.5% bile. Number of genes Number of genes over-expressed under-expressed Gene classification during bile adaptation during bile adaptation Energy production and conversion 0 11 Cell division and envelope biogenesis 0 4 Substrate transport and metabolism6 5 23 Translation, ribosomal structure and biogenesis 0 11 Transcription 2 1 Replication, recombination and repair 0 4 Intracellular trafficking, secretion, and vesicular transport 0 2 Signal transduction mechanisms 0 2 Defense mechanisms 1 0 Unknown functions 9 9 Total 17 67 aGenes were classified based on COG domains found in the protein sequence through a search of the JGI Integrated Microbial Genomes database. bThis class represents two COG categories: cell cycle control, cell division, and chromosome partitioning and cell wall/membrane/envelope biogenesis. cThis class represents multiple COG categories that include transport and metabolism of carbohydrates, amino acids, nucleotides, coenzymes, lipids, inorganic ions, and secondary metabolites. Mutations in three genes, Ir0085, Ir1516, and Ir1864, decrease the ability of cells to survive bile shock. Bile salts have been proposed to cause a wide- range of cellular effects, including cell wall or membrane damage, DNA damage, 59 protein denaturation, oxidative stress, and low intracellular pH (3). Several genes were chosen for mutation based on their proposed functions in adapting to a variety of stresses. Disruptions were created using the pVE6007/pORl28 system in nine genes that were found to be significantly over-expressed during the bile exposure of L. reuteri ATCC 55730 (Table 2.4). Two Clp chaperones (lr0004 [clpE]) and lr1864 [clpL]) were disrupted; Clp chaperones have been implicated in the heat shock response of Bacillus subtilis, as well as other gram- positive organisms (8, 11). The dps gene (lr1706) and a putative esterase (lr1516) were also disrupted to investigate the proposed oxidative stress and cell wall damage effects of bile. The putative esterase (lr1516) belongs to a cluster of orthologous genes (COG) that includes B-Iactamase class C and other various penicillin-binding proteins (20). Three other genes of unknown function were also chosen for disruption: lr1291, a putative metalloproteinase, lr1351, a conserved membrane protein, and Ir0085, a gene of unknown function that appears to be specific to the species L. reuteri. Finally, two multidrug resistance transporters were disrupted (Ir1265 and lr1584). E. coli has the ability to actively pump bile salts out of the cell, and efflux pumps, similar to drug resistance transporters, have been found to have a major role in this activity (37). 60 Table 2.4. Fold gene expression changes in the presence of bile for genes chosen for disruption. Fold- changeb Fold- b , during change Aegessmn bile during bile Gene no. Annotation shock adaptation lr0004 EF421856 Clp chaperone (ClpE) 5 1.4 lr0085 00233699 Hypothetical protein 3.3 2.5 Multidrug resistance protein (ABC Ir1265 EU038268 transporter family) 3.9 1.5 lr1291 AY970991 Metalloproteinase 2.8 1.6 Conserved membrane protein of [H351 00233687 unknown function 3 4.1 Ir1516 D0219970 Putative esterase 7.3 2.8 Multidrug resistance protein (Major lr1584 EU038252 facilitator superfamily) 1.6 2.2 lr1706 EU038255 Dps 2.6 2.1 Ir1864 D0219976 Clp chaperone (CIpL) 3.2 1.4 a . . GenBank accessron numbers are prOVIded. bFold-changes in bold were found to be significantly different based on the outlier analysis. Each of the nine mutants was subjected to treatment with various concentrations of bile to determine which mutants are defective in surviving bile shock. Based on these preliminary experiments, the survival of six of these mutant strains were quantitated and compared to the viability of wild-type cells after 30 minutes of 0.3% bile exposure; the other three mutants (lr0004, lr1291, and lr1351) showed no defect in bile shock and were not further tested. Using a Student’s t-test, it was determined that the survival rate after 30 minutes of bile exposure for three of the mutants (lr1864, lr1516, and lr0085) was significantly different from that of 61 wild-type (p<0.001). Mutations in dps and the two multidrug resistance transporters (lr1706, lr1584, and Ir1265) did not have a significantly different survival rate from wild-type cells (Table 2.5; Figure 2.2). Table 2.5. Effects of 0.3% bile exposure on cell viability and final culture density. Disrupted Accession Percentb gene no.a Annotation survival 0.06000 Ir0085 D0233699 Unknown 16 i 3%* 17:09 Multidrug resistance protein (ABC transporter * Ir1265 EU038268 family) 66 1 23% 1.0301 lr1516 DQZ19970 Putative esterase 8 1 2%* 2110.1 Multidrug resistance protein (Major facilitator * lr1584 EU038252 superfamily) 69 i 32% 0.6100 lr1706 EU038255 Dps 55 i 21% 13:03 lr1864 D0219976 Clp chaperone (CIpL) 3 1 1%* 1.6302 Wild-type cells 54 i 16% 16:02 a . . GenBank acceSSIon numbers are prOVIded. b . . (Percent survwal 30 minutes after exposure. Data is presented as percent survival 1 standard deviation. cData is presented as 0.0500 1 standard deviation after growth had ceased. *p<0.001 compared with wild-type. 62 120 100 1- 80 60 I 40 I " _, 20 * 1: °k m 0 ......4 __.r_. .--.,----. we.-. ._-_..T-,.... 2.... .fi. ATCC PRB190 PR8188 PRB167 PRBI14 PRB126 PRB130 55730 Percent survival Strain Figure 2.2. Comparison of survival after 30 minutes of exposure to 0.3% oxgall for L. reuteri ATCC 55730 wild-type and PRB190 (Ir1864 - CIpL), PRB188 (lr1516 - putative esterase), PRB167 (lr0085 - unknown), PRB114 (lr1706 - Dps), PRB126 (Ir1265 - multidrug resistance protein in the ABC transporter family), and PRB130 (lr1584 - multidrug resistance protein in the major facilitator superfamily) mutant strains. Cultures were plated onto MRS plates after bile exposure to determine the number of viable cells. Error bars represent standard deviation. *p < 0.001 compared with wild-type. Mutations in a putative operon encoding a multidrug resistance protein and a hypothetical protein decrease the ability of L. reuteri to adapt in the presence of bile. When testing the mutant strains for survival in the presence of bile, it was observed that the Ir1584 mutant did not adapt to grow in the presence of bile, even after extended incubations (24 hours). The final culture density 63 obtained for the lr1584 mutant was found to be three-fold lower than that of the wild-type strain. Additional experiments revealed that most of the other mutants were not affected in their ability to adapt and grow in the presence of bile. The exceptions were the Ir1265 mutant, which also obtains a lower culture density than wild-type, and lr1516 mutant, which obtains a slightly higher culture density (Table 2.5). The decreased ability of the Ir1584 and Ir1265 mutant strains to adapt to the presence of bile suggests that multidrug resistance efflux pumps play a role in this strain’s bile response. Efflux pumps have already been shown to play important roles in the bile response of other bacteria (18, 34, 37). The lr1584 gene is found in a putative operon with a gene encoding a conserved hypothetical protein, lr1582. Ir1582 is also found to be significantly over- expressed in the presence of bile; therefore, we were concerned about the possible polar effects the disruption in lr1584 would have on the downstream gene, lr1582. To distinguish between the effects of the Ir1584 mutation and the possible polar effects on Ir1582, a separate mutant strain containing a disruption in Ir1582 was created and tested for its ability to adapt in the presence of bile. This strain also showed an adaptation defect; the final culture density of the Ir1582 mutant was two-fold lower than that of wild-type cells. This demonstrates that the adaptation defect seen in the Ir1584 mutant cannot be fully explained by polar effects on the downstream gene, Ir1582, and suggests that both genes in this operon play a role in L. reuteris adaptation to bile. 64 DISCUSSION The ability of a bacterium to resist bile stress is one of the criteria often used in the selection of a potential probiotic. Bile is a complex mixture of bile acids, phospholipids, proteins, ions, and pigments that has potent antimicrobial properties, particularly against gram-positive bacteria. In this study we have identified several genes that participate in the ability of Lactobacillus reuteri ATCC 55730 to tolerate bile shock and to resume growth in the presence of bile (bile adaptation). Stress responses activated in L. reuteri based on gene expression data. The gene expression data indicate that membrane/cell wall stress, oxidative stress, DNA damage, and protein denaturation occur when L. reuteri is exposed to bile. Several of these pathways have been previously shown to be involved in dealing with various forms of stress in other bacteria. First, the Clp chaperones ClpE and CIpL are induced 15 minutes after bile addition as is their known transcriptional regulator in other gram-positive organisms, CtsR. CtsR is a repressor of multiple clp chaperones in Listeria monocytogenes and Bacillus subtilis and also represses its own expression. Previous work has shown that the induction of the CtsR stress regulon is transient with an initial peak of expression under heat or salt stress that then is reduced after a period of time (32). Consistent with this mode of regulation in other bacteria is the fact that we observe that ctsR and the clp chaperones are overexpressed only during bile 65 shock and not during bile adaptation. c/pL was specifically required for L. reuteri to resist bile shock while the clpE mutant did not survive significantly different than wild-type cells. Repeated attempts to construct a mutation in the ctsR gene were unsuccessful. Second, we also observed increased expression of dps, a protein involved in several types of stress adaptation in Escherichia coli including oxidative stress, irradiation, metal toxicity, heat stress, and pH stress (22, 24). However, disruption of dps in L. reuteri did not significantly affect their ability to survive bile shock or adapt to the presence of bile. Finally, two additional stress response genes and one additional pathway were also induced. A homolog of the gene glsZ4 (lr2108) was induced; G|324 was previously identified as a bile- induced protein in Enterococcus faecalis. Subsequent genetic analysis indicated it was required for the ability of E. faecalis to survive bile exposure, however no molecular function for Gls24 is known (12). The gene lr1346 encodes a homolog of the phage shock transcriptional regulator PspC, which is proposed to be involved in sensing membrane stress during phage infection. Given that bile likely induces membrane stress it is possible that lr1346 plays a role in bile stress survival. Unfortunately we were unable to disrupt lr1346 due to limitations of our gene knockout technology. Lastly, genes of the arginine deiminase pathway were specifically induced during bile adaptation. This pathway has been implicated in the ability to resist mild pH shock in bacteria (21). The identification of lr1516, a putative esterase of the serine B-lactamase-Iike superfamily, as a key enzyme in responding to bile stress suggests these cells 66 are experiencing cell envelope damage upon exposure to bile. Lr1516 contains the signature SxxK active site motif associated with these enzymes, which also include the D-alanyl-D-alanine carboxypeptidases. These enzymes are involved in the breakdown and reorganization of peptidoglycan, and thus we expect that lr1516 may play a similar role when adapting to bile and acid stress (42). Bile adaptation. Because L. reuteri has been shown to colonize, at least temporarily, the small intestine, we were interested in determining if L. reuteri can thrive in the presence of bile. Our results demonstrate that L. reuteri can sustain growth in the presence of bile concentrations as high as 5%. Interestingly the data suggest a multidrug resistance transporter (lr1584) is required for this ability to grow in the presence of bile, suggesting that removal of bile or another toxic metabolite from the cytoplasm is required for growth. Lr1584 is a member for the EmrB/QacA subfamily of the major facilitator superfamily of multidrug resistance transporters. EmrB has previously been shown to play a role in bile resistance and efflux of bile in Escherichia coli. (37). Interestingly, lr1584 is found in an operon upstream of a conserved hypothetical protein, Ir1582; this operon is conserved in many lactic acid bacteria and is over- expressed during exposure of L. reuteri, L. acidophilus, and E. faecalis to bile (25, 35). Due to the limited genetic tools available for use in L. reuteri, we were not completely able to distinguish between the effects of disrupting Ir1584 and possible polar effects this disruption may have on the downstream Ir1582. A 67 separate mutant strain with a disruption in Ir1582 was created and tested for bile adaptation. The Ir1582 mutant does result in an adaptation defect, although it is not as severe as the defect found in the lr1584 mutant strain. The final culture density of the lr1584 mutant is approximately 3-fold lower than that of the wild- type cells, while the final culture density of the Ir1582 mutant is approximately 2- fold lower than wild-type cells. This suggests that both genes may contribute to the adaptation defect that we have observed. Further investigation is required to elucidate the specific role of each gene in bile adaptation. Genes that provide protection in bile stress also protect against acid stress. We identified three genes in L. reuteri that were induced by bile stress and that significantly reduced their ability to survive bile shock when disrupted. Two of these proteins have recently been shown to be induced by a strong reduction in pH and are necessary for increased survival at low pH (42). Both lr1864 (clpL) and lr1516 (putative esterase) play a role in surviving the initial shock of acid and bile stress. Indeed nearly one-third of the genes Wall et al. found to be differentially regulated in acid stress conditions were also altered under bile stress conditions (Table 2.6) (42). This indicates that once cells experience acid stress in the stomach many of the important pathways for dealing with bile stress in the small intestine will already be activated. 68 Table 2.6. Genes over-expressed or under-expressed during both 15 minutes of bile exposure (0.5% oxgall) and 15 minutes of acid stress (pH 2.7) (42). Accession Gene no.8 Annotation Over-expressed lr0597 00219952 Thioredoxin domain-containing protein Ir0922 00074860 Extracellular hydrolase Ir1139 AY970988 Conserved intracellular protein of unknown function lr1191 00219999 Conserved membrane protein of unknown function Ir1468 00219968 Putative transcriptional regulator lr1515 00074905 Unknown extracellular protein lr1516 00219970 Putative esterase lr1797 00219975 Phosphatidylglycerolphosphatase A and related proteins Ir1864 00219976 CIpL ATPase with chaperone activity Ir1937 00219979 Conserved intracellular protein of unknown function lr1993 00219980 Putative transcriptional regulator lr2045 00219981 Phage-associated protein Under-expressed lr0190 00219995 Transcriptional regulator lr0195 00219996 Putative 5-formyltetrahydrofolate cyclo-ligase Ir0382 AY971000 Putative branched-chain amino acid transport protein Ir0733 00219954 Conserved intracellular protein of unknown function lr0862 00219957 Asp-tRNAAsn/Glu-tRNAGIn amidotransferase C subunit Ir1240 00219962 Recombinational DNA repair ATPase Ir1297 00219963 Thymidine kinase lr1432 00220005 Ribosomal protein S1 lr1434 00074902 Unknown extracellular protein Conserved intracellular protein, MarZ, of unknown Ir1628 00219972 function a . . GeneBank acceSSIon numbers are prOVIded. Comparison of multiple genomic studies of bile stress in lactic acid bacteria. Multiple genomic studies have now been completed that identified genes important for bile tolerance in different species of Lactobacillus and Enterococcus (5, 6, 25, 35). Although different culture conditions, types of bile, and species were used for these studies, which resulted in a limited overlap in the genes identified in these studies, there are some common themes that have 69 emerged. In both Lactobacillus plantarum and Lactobacillus acidophilus several genes involved in the reorganization of the cell envelope were induced. Thus dealing with membrane stress is a common theme that has emerged from these three studies. In addition, the operon containing Ir1584 (multidrug resistance transporter) and Ir1582 (unknown function) is also conserved as an operon in L. acidophilus and Enterococcus faecalis. Interestingly, both homologs in E. faecalis and L. acidophilus are also overexpressed when cells were exposed to bile, indicating the function of this operon in bile adaptation may be conserved in other lactic acid bacteria (25, 35). Lastly, clp proteases were identified in L. acidophilus as being upregulated by bile stress as we found with clpL in our study, further supporting that protein denaturation is one stress being encountered by bile treated cells. One common finding that is not easily explained is the reduction in gene expression of recF, which encodes a protein that participates in the repair of DNA damage during active DNA replication. Since bile has been implicated in generating DNA damage, on the surface it seems that a reduction in the expression of RecF would not be productive. However, recent evidence indicates that RecF is predominantly utilized in DNA repair at replication forks during active growth. The reduction in recF expression may simply indicate a reduction in growth rate in the presence of bile. 7O Although there were significant similarities in the transcriptional profiles between the E. faecalis, L. acidophilus, L. plantarum, and L. reuteri responses to bile treatment; overall there was much more discordance in the data than similarities. The lack of concordance may be due to the different physiological strategies utilized by these organisms to adapt to bile stress. In addition, the differences in experimental strategy (the type of bile used, the way bile was administered, and for how long) likely also played a significant role in the differences that were noted. Bron et al. exposed L. plantarum to 0.1% porcine bile on plates and looked at the response to three days of exposure, while Pfeiler et al. conducted their L. acidophilus experiments in liquid media containing 0.5% oxgall with 30 minutes of exposure (6, 25). The use of purified bile acids or different sources of bile will also have different effects on cell physiology (3). Bile has been implicated as a potential signaling molecule that would indicate to a bacterium that it had entered the small intestine. Such a signal could serve to stimulate the organism to adapt its physiology to optimize growth and survival in the GI tract. Several candidates from these experiments have now been identified in vitro. Future work in relevant animal models will determine if the strategies uncovered here are important for survival in vivo. 71 SUPPLEMENTAL INFORMATION Table 2.7. Genes over-expressed after 15 minutes of exposure to 0.5% oxgafl. Accession Fold- Gene name no.a Functional Classificationb change Cell division and envelope biogenesisc ABC-type transport system involved in lipoprotein release permease lr1816 00074937 component 3.4 Defense mechanisms ‘ Multidrug resistance ABC transporter ATP-binding and Ir1265 EU038268 permease protein 3.9 lr1516 00219970 Putative esterase 7.3 ABC transporter, ATP-binding lr1817 EU038262 protein 2.3 Unknown function lr0085 00233699 Hypothetical protein 3.3 Ir0540 EU038232 Conserved hypothetical protein 2.9 lr0542 EU038234 Conserved hypothetical protein 3.1 |r0543 EU038235 Cva family protein 2.3 lr0922 00074860 Extracellular hydrolase 2.2 Conserved membrane protein of lr1 191 00219999 unknown function 3.2 Conserved intracellular protein of lr1 139 AY970988 unknown function 2.5 Intracellular protein of unknown lr1350 00233686 function 5.3 Conserved membrane protein of lr1351 00233687 unknown function 3.1 lr1515 00074905 Unknown extracellular protein 13.6 lr1684 00857799 Conserved hypothetical protein 2.5 lr1740 EU038256 Acetyltransferase (EC 2.3.1 .-) 3.5 Conserved hypothetical protein, lr1755 EU038257 putative pseudogene 2.5 72 Table 2.7 (cont’d) Conserved intracellular protein of Ir1937 00219979 unknown function 2.3 lr1990 00074949 Unknown extracellular protein 2.5 lr1994 EF421933 Conserved hypothetical protein 3.1 lr2045 00219981 Phage-associated protein 2.3 lr2108 00233697 Putative stress response protein 2.6 Posttranslational modification, protein turnover, and chaperones ATP-dependent clp protease ATP- lr0004 EF421856 binding subunit clpE 4.9 Thioredoxin domain-containing lr0597 00219952 protein 2.2 Ir1268 00857779 Hypothetical protein 2.4 lr1291 AY970991 Putative metalloproteinase gene 2.8 lr1788 EU038260 Thioredoxin 2.2 CIpL ATPase with chaperone lr1864 00219976 activity 3.2 Replication, recombination, and repair Holliday junction resolvase-like lr0541 EU038233 protein 3.2 lr0685 EU421874 Transposase 3.3 DNA mismatch repair protein Ir2132 EU038269 MutS2 2.5 Transcription lr0783 EF421875 Transcriptional regulator ctsR 2.3 Transcriptional regulator, MarR lr1264 EU038242 family 3.8 Stress-responsive transcriptional Ir1346 EU038243 regulator PspC 2.7 Transcriptional regulator, TetR lr1347 EU038244 family 4.3 Ir1468 00219968 Putative transcriptional regulator 3.1 lr1573 E7038251 Transcription regulator, Aan-type 2.3 Ir1815 EU038261 Transcriptional regulator 2.5 lr1993 00219980 Putative transcriptional regulator 2.2 73 Table 2.7 (cont’d) Translation, ribosomal structure and biogenesis Ribosome-associated protein Y lr1067 EF421891 (PSrp-1) 2.4 Ribosomal large subunit pseudouridine synthase, RIuD lr1786 E7038259 subfamily 3.1 Substrate transport and metabolismd Transcriptional regulator, GntR lr0621 EU038237 family / aminotransferase 2.2 Thiamine pyrophosphate-requiring lr1 1 17 00857776 enzymes 2.3 DPS (DNA protection during Ir1706 EU038255 starvation) protein 2.6 Phosphatidylglycerolphosphatase A lr1797 00219975 and related proteins 2.5 a GenBank accesion numbers are provided. b Genes were classified based on COG domains found in the protein sequence through a search of the JGI Integrated Microbial Genomes database. ‘ c This class represents two COG categories: cell cycle control, cell division, and chromosome partitioning and cell wall/membrane/envelope biogenesis. d This class represents multiple COG categories that include transport and metabolism of carbohydrates, amino acids, nucleotides, coenzymes, lipids, inorganic ions, and secondary metabolites. 74 Table 2.8. Genes over-expressed during growth in the presence of 0.5% oxgafl. Accession Fold- Gene name no.a Functional Classificationb champ Defense mechanisms lr151 6 00219970 Putative esterase 2.8 Unknown function Ir0085 00233699 Hypothetical protein 2.5 lr0890 00233682 Putative acetyltransferase 2.4 Ir1035 00857774 Conserved hypothetical protein 2.2 lr1348 00074898 predicted membrane protein 2.1 Intracellular protein of unknown lr1350 00233686 function 5.4 Conserved membrane protein of lr1351 00233687 unknown function 4.1 lr1515 00074905 Unknown extracellular protein 3.7 Ir1582 EF421918 Conserved hypothetical protein 2.8 Ir2108 00233697 Putative stress response protein 2.3 Transcription Transcriptional regulator, TetR lr1347 EU038244 family 2.3 lr1518 00233704 ArgR Arginine repressor 2.2 Substrate transport and metabolismc lr1019 00857773 ArcC Carbamate kinase 2.0 ArgF Ornithine lr1020 00233707 carbamoyltransferase 2.7 Ir1517 00233695 ArcA Arginine deiminase 2.6 Permease of the major facilitator lr1584 EU038252 superfamily 2.2 DPS (DNA protection during Ir1706 EU038255 starvation) protein 2.1 a GenBank accesion numbers are provided. b Genes were classified based on COG domains found in the protein sequence through a search of the JGI Integrated Microbial Genomes database. c This class represents multiple COG categories that include transport and metabolism of carbohydrates, amino acids, nucleotides, coenzymes, lipids, inorganic ions, and secondary metabolites 75 Table 2.9. Genes under-expressed after 15 minutes of exposure to 0.5% oxgafl. ACCESSION FOId- Gene name no.a Functional Classificationb change . . . . C Cell leISIOI‘I and envelope biogenesis lr0548 EU038236 Cell division initiation protein 3.2 Capsular polysaccharide lr0957 00857880 biosynthesis protein 1.9 S-adenosyl-methyltransferase lr1629 EU038253 MraW 2 .4 Energy production and conversion CitC Citrate lyase synthetase lr0599 00233700 partial CDS 1.9 lr0600 00240820 Sch malic enzyme 2.1 lr1073 EF534266 Fumarase 2.0 PduA propanediol utilization protein: putative lr1882 00233726 microcompartment protein 2.9 Unknown function lr0196 EU038226 Rhomboid family protein 1.9 Conserved intracellular protein of lr0733 00219954 unknown function 2.7 lr1241 EF421898 Conserved hypothetical protein 3.0 lr1267 00074894 Unknown extracellular protein 2.1 lr1434 00074902 Unknown extracellular protein 2.0 Conserved intracellular protein, Ir1628 00219972 MarZ, of unknown function 2.2 lr2134 EU038270 ComG operon protein 1 2.4 Posttranslational modification, protein turnover, and chaperones lr1373 EU038246 FeS assembly ATPase Squ 1.9 lr1774 EU038258 Glutaredoxin-like protein NrdH 3.0 Replication, recombination, and repair ATPase related to the helicase subunit of the Holliday junction lr0044 EF421 958 resolvase 2.3 Nuclease subunit of the lr0311 00857869 excinuclease complex 2.0 Recombinational DNA repair lr1240 00219962 ATPase 3.4 76 Table 2.9 (cont’d) Transcription lr0190 00219995 predicted transcriptional regulator Translation, ribosomal structure and biogenesis Asp-tRNAAsn/Glu-tRNAGIn lr0862 00219957 amidotransferase C subunit 2.6 lr1432 00220005 Ribosomal protein S1 2.4 Substrate transport and metabolismd Spermidine/putrescine ABC lr0137 EF537897 transporter, permase protein 3.4 lr0160 EF547651 Sugar kinase, ribokinase family 4.3 lr0187 EU038223 Dihydroneopterin aldolase 2.3 2-amino-4-hydroxy-6- hydroxymethyldihydropteridine lr0188 EU038224 pyrophosphokinase 2.0 lr0189 EU038225 GTP cyclohydrolase l 1.9 Putative 5-formyltetrahydrofolate lr0195 00219996 cycIo-ligase 2.2 Cystathionine beta-lyase (EC 4.4.1.8) / Cystathionine gamma- lr0324 EF421866 lyase (EC 4.4.1.1) 2.9 Putative branched-chain amino lr0382 AY971000 acid transport protein 2.1 Permease of the major facilitator Ir0848 EU038267 superfamily 2.1 Ir1297 00219963 Thymidine kinase 2.1 PduC propanediol dehydratase lr1880 00233724 large subunit 2.0 , PduB propanediol utilization Ir1881 00233725 protein 3.1 lr1959 00857828 HemA, Glutamyl-tRNA reductase 2.2 lr1960 00857829 Putative siroheme synthase 2.7 lr1961 00857830 CbiP, cobyric acid synthase 2.3 |r1963 00857832 Cbi0, cobalt transport protein 2.2 CbiB, cobalamin biosynthesis lr1977 00857846 protein 2.0 CbiA, cobyrinic acid a,c-diamide lr1978 00857847 synthase 2.2 CobD, L-threonine-0-3-phosphate Ir1979 00857848 decarboxylase 3.2 77 Table 2.9 (cont’d) Diacylglycerol kinase family lr2131 EU038263 protein 2.4 ABC-type ribose transport system, lr2133 EU038271 auxiliary component 2.3 a GenBank accession numbers are provided. b Genes were classified based on COG domains found in the protein sequence through a search of the JGI Integrated Microbial Genomes database. c This class represents two COG categories: cell cycle control, cell division, and chromosome partitioning and cell wall/membrane/envelope biogenesis. d This class represents multiple COG categories that include transport and metabolism of carbohydrates, amino acids, nucleotides, coenzymes, lipids, inorganic ions, and secondary metabolites. 78 Table 2.10. Genes under-expressed during growth in the presence of 0.5% oxgall. Accession Fold- Gene name no.a Functional Classificationb change Cell division and envelope biggenesisc GidB predicted S- adenosylmethionine-dependent Ir0145 00219992 methyltransferase partial CDS 1.7 WcaA Glycosyltransferases lr0740 00233678 involved in cell wall biogenesis 1.6 Capsular polysaccharide lr0957 00857880 biosynthesis protein 1.7 UDP-MurNAc—pentapeptide lr1645 EU038254 synthetase 1 .8 Energy production and conversion CitC Citrate lyase synthetase lr0599 00233700 partial CDS 2.1 lr0600 00240820 Sch malic enzyme 1.7 Fumarate reductase fiavoprotein lr1074 EF534267 subunit (EC 1.3.99.1) 2.5 L-lactate dehydrogenase (EC lr1075 EF421892 1.1.1.27) 2.8 Cytochrome bd-type quinol lr1236 EF534271 oxidase, subunit 1 1.9 Ir1402 EF421914 ATP synthase subunit A 1.7 Ir1403 EF421915 ATP synthase subunit C 1.6 lr1404 EU038247 ATP synthase F0, B subunit 1.8 lr1405 EU038248 ATP synthase F1, delta subunit 1.7 lr1406 EU038249 ATP synthase F1, alpha subunit 1.7 COG3051 CitF Citrate lyase lr1418 00233691 alpha subunit 1.9 Intracellular trafficking, secretion, and vesicular transport COG0706 preprotein translocase lr0252 00074813 subunit YidC 1 .6 COG0201 preprotein translocase Ir0469 00074824 subunit SecY 1.8 79 Table 2.10 (cont’d) Unknown function Beta-phosphoglucomutase (EC 5.4.2.6) / GIucose-1-phosphate 80 lr1054 00466579 phosphodismutase (EC 2.7.1.41) 1.6 lr1487 00074904 Unknown extracellular protein 1.7 Ir2054 00219984 Phage head maturation protease 2.0 lr2057 00219986 Putative phage protein 1.7 lr2058 00219987 Putative phage protein 1.6 Ir2134 EU038270 ComG operon protein 1 2.5 lr2135 EU038264 Membrane protein, putative 1.5 lr2136 EU038265 Hypothetical protein 1.6 lr2137 EU038266 Amino acid transporter 1.6 Replication, recombination, and repair Chromosomal replication initiator lr0138 EU038222 protein DnaA 1.6 Nuclease subunit of the lr0311 00857869 excinuclease complex 1.7 Ir1238 EU038240 DNA gyrase, A subunit 1.8 Ir1239 EU038241 DNA gyrase, B subunit 1.6 jignal transduction mechanisms . ' Stress response membrane lr0279 EF421950 GTPase TypA 1.6 Transcriptional regulator, MarR lr1012 EF421886 family 1.8 Transcription DNA-directed RNA polymerase, lr1358 EU038245 beta subunit 1.6 Translation, ribosomal structure and biogenesis lr0050 EU038217 Ribosomal protein L14 2.3 lr0051 EU038218 Ribosomal protein S17 2.2 lr0062 EF421960 Ribosomal protein S8 2.1 Ir0063 EU038219 Ribosomal protein L6 2.0 lr0251 00219950 Rnase P protein component 1.7 Ir0286 EU038227 Ribosomal protein S13p 1.9 lr0287 EU038228 Ribosomal protein S11 2.0 lr0470 EF421870 Ribosomal protein L15 1.9 Ir0471 EU038230 Ribosomal protein L30 1.9 lr0472 EU038231 Ribosomal protein 35 1.9 TetW, Tetracycline resistance lr1996 EF421935 protein 1.8 Table 2.10 (cont’d) Substrate transport and metabolismd lr0092 Ir0101 Ir0136 lr0137 lr0160 Ir0324 #0467 00673 k0674 h1009 Ir1013 "1198 k1287 k1417 01419 ”1508 "1961 "1963 01964 01967 EF421961 EF421948 EU038221 EF537897 EF547651 EF421866 EU038229 EU038238 EU038239 EF421883 EF421887 00074887 EF421 909 D0233690 00233692 EU038250 D0857830 00857832 00857833 DQ857836 Deoxyribose-phosphate aldolase (EC 4.1.2.4) Phosphopentomutase (EC 5.4.2.7) Spermidine/putrescine ABC transporter, permease protein Spermidine/putrescine ABC transporter, permase protein Sugar kinase, ribokinase family Cystathionine beta-lyase (EC 4.4.1.8) / Cystathionine gamma- lyase (EC 4.4.1.1) Adenylate kinase (ATP-AMP transphosphorylase) Purine nucleoside phosphorylase Pyrimidine-nucleoside phosphorylase (acyl-carrier-protein) S- malonyltransferase, FabD 3-hydroxymyristoyl/3- hydroxydecanoyl-(acyl carrier protein) dehydratase, FabA HisJ ABC-type amino acid transport/signal transduction systems periplasmic component/HisM ABC-type amino acid transport system permease component 3-oxoacyl-(acyl-carrier—protein) synthase, FabB Triphosphoribosyl-dephospho- COA synthetase Citrate lyase beta subunit Permease, GntP family CbiP, cobyric acid synthase Cbi0, cobalt transport protein CbiN, cobalt transport protein CbiK, cobalt chelatase 81 3.1 2.9 1.6 1.8 2.3 2.1 1.8 1.7 2.9 1.6 1.8 1.9 1.7 1.9 1.8 1.7 1.7 1.6 1.5 1.8 Table 2.10 (cont’d) CbiH, precorrin-3B C17- lr1970 00857839 methyltransferase 1 .6 lr2131 EU038263 Transcription regulator 2.0 lr2133 EU038271 Ribose transport protein 2.8 a GenBank accesion numbers are provided. b Genes were classified based on COG domains found in the protein sequence through a search of the JGI Integrated Microbial Genomes database. c This class represents two COG categories: cell cycle control, cell division, and chromosome partitioning and cell wall/membranelenvelope biogenesis. d This class represents multiple COG categories that include transport and metabolism of carbohydrates, amino acids, nucleotides, coenzymes, lipids, inorganic ions, and secondary metabolites. Acknowledgements. We would like to thank Eamonn Connolly for helpful discussions and access to the L. reuteri ATCC 5730 genome and Eric Hufner and Christian Hertel for assistance in generating mutants. We also thank Jeff Landgraf in the Research Technology Support Facility at Michigan State University for assistance with microarray production. This work was supported in part by funding support to RAB from the Michigan State Center for Microbial Pathogenesis and the Rackham Foundation. 82 10. REFERENCES Bath, K., S. Roos, T. Wall, and H. Jonsson. 2005. The cell surface of Lactobacillus reuteri ATCC 55730 highlighted by identification of 126 extracellular proteins from the genome sequence. FEMS Microbiol Lett 253:75-82. Begley, M., C. G. Gahan, and C. Hill. 2002. Bile stress response in Listeria monocytogenes L028: adaptation, cross-protection, and identification of genetic loci involved in bile resistance. Appl. Environ. Microbiol. 68:6005-12. Begley, M., C. G. Gahan, and C. Hill. 2005. The interaction between bacteria and bile. FEMS Microbiol. Rev. 29:625-51. Britton, R. A., P. Eichenberger, J. E. Gonzalez-Pastor, P. Fawcett, R. Monson, R. Losick, and A. D. Grossman. 2002. Genome-wide analysis of the stationary-phase sigma factor (sigma-H) regulon of Bacillus subtilis. J. Bacteriol. 184:4881-90. Bron, P. A., M. Marco, S. M. Hoffer, E. Van Mullekom, W. M. de Vos, and M. Kleerebezem. 2004. Genetic characterization of the bile salt response in Lactobacillus plantarum and analysis of responsive promoters in vitro and in situ in the gastrointestinal tract. J. Bacteriol. 186:7829-35. Bron, P. A., D. Molenaar, W. M. de Vos, and M. Kleerebezem. 2006. DNA micro-array-based identification of bile-responsive genes in Lactobacillus plantarum. J. Appl. Microbiol. 100:728-738. Casas, I., and W. Dobrogosz. 2000. Validation of the probiotic concept: Lactobacillus reuteri confers broad-spectrum protection against disease in humans and animals. Microb. Ecol. Health Dis. 12:247-285. Derré, I., G. Rapoport, K. Devine, M. Rose, and T. Msadek. 1999. ClpE, a novel type of HSP100 ATPase, is part of the CtsR heat shock regulon of Bacillus subtilis. Mol. Microbiol. 32:581-93. Dunne, C., L. Murphy, S. Flynn, L. O'Mahony, S. O'Halloran, M. Feeney, D. Morrissey, G. Thornton, G. Fitzgerald, C. Daly, B. Kiely, E. M. Quigley, G. C. O'Sullivan, F. Shanahan, and J. K. Collins. 1999. Probiotics: from myth to reality. Demonstration of functionality in animal models of disease and in human clinical trials. Antonie Leeuwenhoek 76:279-92. FAO. 2001. Health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria: report of a joint FAO/WHO 83 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. expert consultation on evaluation of health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria. Food and Agriculture Organization. Frees, D., K. Savijoki, P. Varmanen, and H. lngmer. 2007. Clp ATPases and ClpP proteolytic complexes regulate vital biological processes in low GC, Gram-positive bacteria. Mol. Microbiol. 63:1285-95. Giard, J. C., A. Rince, H. Capiaux, Y. Auffray, and A. Hartke. 2000. Inactivation of the stress- and starvation-inducible gls24 operon has a pleiotrophic effect on cell morphology, stress sensitivity, and gene expression in Enterococcus faecalis. J. Bacteriol. 182:4512-20. Gunn, J. S. 2000. Mechanisms of bacterial resistance and response to bile. Microbes Infect. 2:907-13. Hofman, A. 1998. Bile secretion and the enterohepatic circulation of bile acids., p. 937-948. In M. Feldman, B. Scharschmidt, and M. Sleisenger (ed. ), Sleisengers and Fordtran' s Gastrointestinal and Liver Disease, 6 ed. W. B. Saunders, Co., Philadelphia. Law, J., G. Buist, A. Haandrikman, J. Kok, G. Venema, and K. Leenhouts. 1995. A system to generate chromosomal mutations in Lactococcus lactis which allows fast analysis of targeted genes. J. Bacteriol. 177:7011-8. Le Breton, Y., A. Mazé, A. Hartke, S. Lemarinier, Y. Auffray, and A. Rincé. 2002. Isolation and characterization of bile salts-sensitive mutants of Enterococcus faecalis. Curr. Microbiol. 45:434-9. Leverrier, P., D. Dimova, V. Pichereau, Y. Auffray, P. Boyaval, and G. Jan. 2003. Susceptibility and adaptive response to bile salts in Propionibacterium freudenreichii: physiological and proteomic analysis. Appl. Environ. Microbiol. 69:3809-18. Lin, J., 0. Sahin, L. 0. Michel, and 0. Zhang. 2003. Critical role of multidrug efflux pump CmeABC in bile resistance and in vivo colonization of Campy/obacterjejuni. Infect. Immun. 71 :4250-9. Maguin, E., P. Duwat, T. Hege, D. Ehrlich, and A. Gruss. 1992. New thermosensitive plasmid for gram-positive bacteria. J. Bacteriol. 174:5633- 8. Marchler-Bauer, A., and S. H. Bryant. 2004. CD-Search: protein domain annotations on the fly. Nucleic Acids Res. 32:W327-31. 84 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. Marquis, R. E., G. R. Bender, D. R. Murray, and A. Wong. 1987. Arginine deiminase system and bacterial adaptation to acid environments. Appl. Environ. Microbiol. 53:198-200. Martinez, A., and R. Kolter. 1997. Protection of DNA during oxidative stress by the nonspecific DNA-binding protein Dps. J. Bacteriol. 179:5188- 94. Metchnikoff, E. 1907. Lactic acid as inhibiting intestinal putrefaction., p. 161-183, The prolongation of life: optimistic studies. W. Heinemann, London. Nair, S., and S. E. Finkel. 2004. Dps protects cells against multiple stresses during stationary phase. J. Bacteriol. 186:4192-8. Pfeiler, E. A., M. A. Azcarate-Peril, and T. R. Klaenhammer. 2007. Characterization of a novel bile-inducible operon encoding a two- component regulatory system in Lactobacillus acidophilus. J. Bacteriol. 189:4624-34. Reid, G. 1999. The scientific basis for probiotic strains of Lactobacillus. Appl. Environ. Microbiol. 65:3763-6. Reuter, G. 2001. The Lactobacillus and Bifidobacterium microflora of the human intestine: composition and succession. Curr. Issues lntest. Microbiol. 2:43-53. Rolfe, R. D. 2000. The role of probiotic cultures in the control of gastrointestinal health. J. Nutr. 130:396S-402S. Russell, W. M., and T. R. Klaenhammer. 2001. Efficient system for directed integration into the Lactobacillus acidophilus and Lactobacillus gasseri chromosomes via homologous recombination. Appl. Environ. Microbiol. 67:4361-4. Savino, F., E. Pelle, E. Palumeri, R. Oggero, and R. Miniero. 2007. Lactobacillus reuteri (American Type Culture Collection Strain 55730) versus Simethicone in the treatment of infantile colic: a prospective randomized study. Pediatrics 119:9124-e130. Saxelin, M., S. Tynkkynen, T. MattiIa-Sandholm, and W. M. de Vos. 2005. Probiotic and other functional microbes: from markets to mechanisms. Curr. Opin. Biotechnol. 16:204-11. Schumann, W., M. Hecker, and T. Msadek. 2002. Regulation and Function of Heat-Inducible Genes in Bacillus subtilis, p. 359-368. In A. L. 85 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. Sonenshein, J. A. Hoch, and R. Losick (ed.), Bacillus subtilis and It's Closest Relatives: From Genes to Cells. ASM Press, Washington, DC. Simons, L. A., S. G. Amansec, and P. Conway. 2006. Effect of Lactobacillus fermentum on serum lipids in subjects with elevated serum cholesterol. Nutr. Metabol. Cardiovasc. Dis. 16:531-5. Sleator, R. D., H. H. Wemekamp-Kamphuis, C. G. Gahan, T. Abee, and C. Hill. 2005. A PrfA-regulated bile exclusion system (BiIE) is a novel virulence factor in Listeria monocytogenes. Mol. Microbiol. 55:1183-95. Solheim, M., A. Aakra, H. Vebo, L. Snipen, and I. F. Nes. 2007. Transcriptional Responses of Enterococcus faecalis V583 to Bovine Bile and Sodium Dodecyl Sulfate. Appl. Environ. Microbiol. 73:5767-74. Talarico, T. L., I. A. Casas, T. C. Chung, and W. J. Dobrogosz. 1988. Production and isolation of reuterin, a growth inhibitor produced by Lactobacillus reuteri. Antimicrob. Agents Chemother. 32:1854-8. Thanassi, D. G., L. W. Cheng, and H. Nikaido. 1997. Active efflux of bile salts by Escherichia coli. J. Bacteriol. 179:2512-8. Tubelius, P., V. Stan, and A. Zachrisson. 2005. Increasing work-place healthiness with the probiotic Lactobacillus reuteri: a randomised, double- blind placebo-controlled study. Environ. Health 4:25. Uicker, W. C., L. Schaefer, and R. A. Britton. 2006. The essential GTPase RbgA (quF) is required for 508 ribosome assembly in Bacillus subtilis. Mol. Microbiol. 59:528-40. Usman, and A. Hosono. 2000. Effect of administration of Lactobacillus gassen' on serum lipids and fecal steroids in hypercholesterolemic rats. J. Dairy Sci. 83:1705-11. Valeur, N., P. Engel, N. Carbajal, E. Connolly, and K. Ladefoged. 2004. Colonization and immunomodulation by Lactobacillus reuteri ATCC 55730 in the human gastrointestinal tract. Appl. Environ. Microbiol. 70:1176-81. Wall, T., K. Bath, R. A. Britton, I-l. Jonsson, J. Versalovic, and S. Roos. 2007. The early response to acid shock in Lactobacillus reuteri involves the CIpL chaperone and a putative cell wall-altering esterase. Appl. Environ. Microbiol. 73:3924-35. Wall, T., S. Roos, K. Jacobsson, A. Rosander, and H. Jonsson. 2003. Phage display reveals 52 novel extracellular and transmembrane proteins from Lactobacillus reuteri DSM 20016(T). Microbiology 149:3493-505. 86 44. 45. 46. Walter, J., P. Chagnaud, G. W. Tannock, D. M. Loach, F. Dal Bello, H. F. Jenkinson, W. P. Hammes, and C. Hertel. 2005. A high-molecular- mass surface protein (Lsp) and methionine sulfoxide reductase B (MsrB) contribute to the ecological performance of Lactobacillus reuteri in the murine gut. Appl. Environ. Microbiol. 71 :979-86. Weizman, Z., G. Asli, and A. Alsheikh. 2005. Effect of a probiotic infant formula on infections in child care centers: comparison of two probiotic agents. Pediatrics 115:5-9. Wenus, C., R. Gail, E. B. Loken, A. S. Biong, D. S. Halvorsen, and J. Florholmen. 14 March 2007. Prevention of antibiotic-associated diarrhoea by a fermented probiotic milk drink. Eur. J. Clin. Nutr. doi:10.1038/sj.ejcn.1602718. 87 CHAPTER 3 CHARACTERIZATION OF THE BILE SALT HYDROLASE ACTIVITY OF LACTOBA CILLUS REUTERI ATCC PTA 6475 INTRODUCTION The gastrointestinal microbiota plays an important role in the normal circulation and excretion of human bile. The primary bile acids, cholate and chenodeoxycholate, are synthesized in the liver from cholesterol. Before secretion into the small intestine, these bile acids are conjugated to either a glycine or taurine molecule, which improves the solubility and detergent properties of these molecules (18). As bile passes through the small intestine into the colon, the bile acids that are not reabsorbed in the ileum are modified by gastrointestinal bacteria. The first step in this modification is removal of the taurine or glycine molecule through the activity of bile salt hydrolase enzymes. This modification then allows for further alterations such as those carried out by bacteria with 7a-dehydroxylation activity. This removal of a hydroxyl group converts cholate and chenodeoxycholate into the secondary bile acids, deoxycholate and lithocholate, respectively (2). Bile salt hydrolase activity has been identified in a wide range of commensal gastrointestinal microorganisms, including both bacteria and archaea (19), and it 88 is often suggested that bile salt hydrolase activity contributes to successful persistence in and colonization of the host gastrointestinal tract. Tanaka et al (27) examined 300 strains of lactic acid bacteria and found a strong correlation between the environment from which the organisms were isolated and the presence of bile salt hydrolase activity. The presence of bile salt hydrolase activity in most intestinal or fecal isolates, and the absence of the activity from strains isolated from milk or vegetable products suggests that there may be a selective advantage in the gastrointestinal tract for strains with deconjugation activity. Indeed, several studies have suggested that bile salt hydrolase activity can contribute to persistence in or colonization of the gastrointestinal tract (4, 11, 13, 19). It is important to note that other studies have not demonstrated this effect. Bateup et al (1) investigated five different strains of lactobacilli in regards to their ability to deconjugate bile acids and colonize the murine gastrointestinal tract. Although they demonstrated a range of bile salt hydrolase activity between the strains (from high levels of activity to no activity), all five strains colonized different locations in the gastrointestinal tract equally well. A separate study in Lactobacillus johnsonii showed that even disrupting all three bile salt hydrolase genes in this organism had no effect on gastrointestinal persistence as measured by fecal culturing (12). Although the activity has been primarily studied in commensal or probiotic organisms such as Lactobacillus spp., Bifidobacterium spp., and Enterococcus spp. (3), recent work has suggested that this activity may act as a novel virulence 89 factor for some pathogens. Research in the pathogenic Listeria monocytogenes has revealed that the ability to deconjugate bile acids is not only controlled by PrfA, a transcriptional regulator known to control other virulence factors, but that the activity also plays an important role in the infection capabilities of this pathogen (4, 13). Despite the fact that bile salt hydrolase activity is commonly found in gastrointestinal microorganisms and is believed to contribute to persistence of commensals and some pathogens in the gastrointestinal tract, the actual purpose of this activity is yet to be determined. Possibilities include: detoxification of conjugated bile acids, contribution to gastrointestinal persistence, nutritional benefit through utilization of the cleaved amino acid moiety, or induction of alterations in the cell membrane that may make that bacteria more resistant to other stresses encountered in the gastrointestinal tract such as defensins (3). An understanding of the function of bile salt hydrolase activity in regards to the bacteria is necessary as the activity also has important consequences on the host. Many probiotic strains exhibit the ability to deconjugate bile acids, and several of these strains are being investigated for the ability to lower serum cholesterol levels in the host. It has been proposed that administration of bacteria with bile salt hydrolase activity may lower cholesterol levels in two ways. The first proposed mechanism is that cholesterol may co-precipitate with deconjugated bile acids, thus causing a higher excretion of cholesterol. The second is that administration of bacteria with this activity may increase the 90 amount of deconjugated bile acids that are excreted, thus increasing the amount of bile acids that need to be synthesized from cholesterol (22, 31, 32). Despite this proposed beneficial effect, various studies have demonstrated that an excess of bacteria with bile salt hydrolase activity in the gastrointestinal tract may lead to abnormal digestive functions, gallstone formation, and possibly a higher tendency towards colon cancer (3). Therefore, a thorough understanding of the role that bile salt hydrolase activity plays regarding both the bacteria and the host is needed. MATERIALS AND METHODS Strains and media. The strains and vectors used in this study are listed in Table 3.1. L. reuteri strains were grown in MRS medium (Dich). PRB240 was grown in MRS containing 10 ug/mL erythromycin. Individual bile acids, as well as ox gall/bovine bile, were ordered from Sigma. 91 Table 3.1. Bacterial strains and plasmids used for this study. Reference or Strains or plasmids Description source Strains Strain containing a chromosomal copy of the E. coli EC1000 pWV01 repA gene; Kanr (21) Biogaia, AB, L. reuteri ATCC 55730 Human breast milk isolate Sweden L. reuteri ATCC PTA Biogaia, AB, 6475 Human breast milk isolate Sweden L. reuteri ATCC PTA 6475 bile salt hydrolase (NT01LR0487) PRBZ40 mutant This study Plasmids Cmr repA-positive temperature-sensitive pVE6007 derivative of pWV01 (23) Emr repA-negative derivative pORl28 of pWV01 (21) pORl28 + 289 bp insert from pKW08 NT01LR0487 This study Phenotypic plate test to determine bile salt hydrolase activity. The initial method used to determine the presence or absence of bile salt hydrolase activity for a particular strain was the plate method developed by Dashkevicz and Feighner (8). For this assay, MRS agar (Difco) was combined with 0.05 to 0.5% (w/v) of individual bile acids. The mixture was autoclaved for 20 minutes before dividing into plates. After the plates solidified, bacterial strains were struck from MRS plates onto the MRS plus 0.05 to 0.5% bile acid plates. Plates were incubated anaerobically for 72 hours at 37°C. Strains were considered to be 92 positive for bile salt hydrolase activity based on either precipitation of the bile acids around bacterial colonies or a change in colony morphology to white opaque matte colonies as previously described (8). Strains were assayed for the ability to deconjugate six conjugated bile acids: taurocholate (TCA), glycocholate (GCA), taurodeoxycholate (TDCA), glycodeoxycholate (GDCA), taurochenodeoxycholate (TCDCA), and glycochenodeoxycholate (GCDCA). TLC to determine bile salt hydrolase activity. In addition to the phenotypic plate assay, thin-layer chromatography (TLC) was used to further investigate the ability of the strains to deconjugate the six bile acids mentioned above. For these assays, MRS broth was combined with 1 mM of the various conjugated bile acids listed above. The mixture was autoclaved, allowed to cool, and left in an anaerobic chamber overnight to equilibrate before inoculation with bacterial cultures. For inoculation, 100 pl of an overnight culture of L. reuteri grown in MRS broth was added to 10 mL of MRS plus 1 mM bile acid. These cultures were then incubated anaerobically at 37°C for up to 72 hours. Cultures were assayed for bile salt hydrolase activity at either 24 or 72 hours after inoculation. For these assays, PRB240 was grown in the presence of 10 ug/mL erythromycin. To test for bile salt hydrolase activity, 5 mL of each culture were removed. The mixture was acidified by adding an equal volume of 1 N HCI. Bile acids were then extracted with 10 mL of ethyl acetate, and the phases were allowed to separate (separation of the two phases was aided by the addition of 93 approximately one mL of a saturated brine solution). The water layer was removed, and the organic layer was washed with 10 mL of a saturated brine solution. After the phases separated, the organic layer (containing the bile salts) was collected and dried over sodium sulfate. The samples were then concentrated to dryness by rotary evaporation. For TLC, the extracted bile acids were resuspended in approximately 0.5 mL of methanol and spotted onto silica gel 60 F254 aluminum-backed plates, along with ' control bile acids (dilute control bile acid solutions contained purified bile acids dissolved in methanol). The TLC plates were run using the following solvent system: cyclohexanezethyl acetatezacetic acid (7:23:3 v/v) as previously described (20). Using this solvent system, deoxycholate and chenodeoxycholate migrate at the same rate. A solvent system that allowed for sufficient separation of these two compounds was not identified. Plates were stained using 33 mM phosphomolybdic acid dissolved in 95% ethanol and heated to visualize spots. Cultures were considered positive for bile salt hydrolase activity based on the appearance of spots representing deconjugated bile acids on the TLC plate. Construction of PRBZ40 (L. reuteri ATCC PTA 6475 with disruption in NT01LR0487). The following concentrations of antibiotics were used: 40 ug/mL kanamycin, 10 ug/mL chloramphenicol, 10 ug/mL erythromycin (L. reuteri) or 400 ug/mL erythromycin (E. coli). The NT01LR0487 (bile salt hydrolase) mutant was created using the system developed by Russell and Klaenhammer (26) with 94 modifications made for use in Lactobacillus reuteri (33). In short, 289 bp from NT01LR0487 from L. reuteri ATCC PTA 6475 was cloned into pORl28. This construct was then transformed into E. coli EC1000, and the transformed cells were grown in LB broth in the presence of erythromycin and kanamycin. pORl28 containing the NT01LR0487 insert was then extracted and transformed into L. reuteri ATCC PTA 6475 containing pVE6007. L. reuteri cells containing both plasmids were grown in MRS broth containing chloramphenicol and erythromycin at the permissive temperature of 35°C for 18 hours. The cultures were then shifted to the non-permissive temperature of 45°C and grown in the presence of erythromycin only. After several passages at 45°C to ensure loss of pVE6007, individual colonies were screened for integration of pORl28 at the desired location by PCR. Colonies were screened for the presence of both flanking regions (using one primer that anneals to pORl28 and One primer that anneals to the chromosome outside of the region cloned into pORl28) and the absence of a correctly-sized wild-type gene. Growth studies to determine the effect of loss of BSH activity. Growth studies of L. reuteri ATCC PTA 6475 and PRB240 in the presence a synthetic “human” bile acid mixture (SHB) were conducted. SHB was made to mimic the bile acid concentrations in the small intestine (5, 15, 24) and contained the following components: 0.46 mM taurocholate, 0.93 mM glycocholate, 0.46 mM taurochenodeoxycholate, 0.93 mM glycochenodeoxycholate, 0.32 mM 95 taurodeoxycholate, and 0.64 mM glycodeoxycholate dissolved in MRS broth. This mixture was autoclaved, cooled, and then injected into air-tight bottles containing an anaerobic atmosphere. Overnight cultures of L. reuteri were used to inoculate the bottles at a starting 0.0.500 = 0.02. Growth was carried out at 37° C with slow shaking. Growth curves were performed in duplicate. Samples for TLC analysis were collected at 1, 4, 7, 10, and 23 hours of growth. In order to investigate the effect of pH on the growth of L. reuteri ATCC PTA 6475 and PR8240 in the presence of bile, growth experiments were performed as described above, but with the synthetic human bile mixture dissolved into a buffered version of MRS broth. For this buffered MRS, the medium was made from the various components: 10 g/L proteose peptone no. 3, 10 g/L beef extract, 5 g/L yeast extract, 20 g/L dextrose, 1 g/L Tween 80, 2 g/L ammonium citrate, dibasic, 0.1 g/L magnesium sulfate, 0.05 g/L manganese sulfate, and 2 g/L dipotassium phosphate (sodium acetate was removed). The medium was then buffered with 200 mM MES, and the pH was adjusted to 7.0. For control experiments, MRS without sodium acetate was made, and the pH was adjusted to 7.0. Both types of medium were autoclaved for 20 minutes before use. Samples for TLC analysis were collected from growth curves at 7 hours and 24 hours of growth and frozen at -20° C until use. pH was also monitored throughout the experiments with the use of pH strips. Growth curves were performed in triplicate. 96 RESULTS L. reuteri ATCC PTA 6475 contains a single bile salt hydrolase that has the ability to deconjugate the six major human bile acids. Previous work in a variety of bacteria has demonstrated that deconjugation of bile acids is due to the activity of one or more enzymes classified as choloylglycine hydrolases (EC 3.5.1.24). These bile salt hydrolase enzymes are responsible for the removal of the glycine or taurine moiety from the conjugated bile acids. A search of the L. reuteri ATCC PTA 6475 genome revealed a single gene, NT01LR0487, with significant similarity to known bile salt hydrolase enzymes. A disruption in the gene was created by insertion of pORl28 as described in the Materials and Methods section. The resulting strain was titled PR8240. The ability of this strain to deconjugate an assortment of bile acids was compared to that of the wild-type strain, L. reuteri ATCC PTA 6475, to determine the effect of this gene disruption. Initial testing to determine the scope of activity for L. reuteri ATCC PTA 6475 was performed using the plate test developed by Dashkevicz and Feighner (1989). L. reuteri ATCC PTA 6475 was plated onto MRS plates containing between 0.05 to 0.5% taurocholate (TCA), glycocholate (GCA), taurodeoxycholate (TDCA), glycodeoxycholate (GDCA), taurochenodeoxycholate (TCDCA), or glycochenodeoxycholate (GCDCA) and incubated anaerobically for up to 72 97 hours. Bile salt hydrolase activity was observed by the precipitation of bile acids surrounding individual colonies of bacteria or an opaque granular white appearance of the colonies as shown in Figure 3.1 and as reported previously (8). Based on these phenotypes, the plate tests revealed that the strain could deconjugate all six of the major bile acids found in human bile. 98 '. '1 Figure 3.1. Different phenotypes observed using the plate test to determine bile salt hydrolase activity. Strains of L. reuten' were plated onto MRS plates with or without bile acids to determine bile salt hydrolase ability. A. L. reuteri ATCC PTA 6475 and the bile salt hydrolase mutant strain, PRB240, plated onto MRS plates containing 0.5% oxgall. Precipitation of bile is observed from ATCC PTA 6475 as halos around the colonies; no activity is observed for PRB240. This phenotype was also observed for plates containing glycodeoxycholate (GDCA) and glycochenodeoxycholate (GCDCA) (data not shown). B. The two strains (ATCC PTA 6475 and PRB240) plated onto MRS plates containing 0.01% taurochenodeoxycholate (TCDCA). Bile salt hydrolase activity for this bile acid was demonstrated by a cloudiness surrounding the colonies that could be observed when the plates were held against a light. No activity is observed for PRBZ40. C. ATCC PTA 6475 plated onto MRS or MRS plus 0.5% taurodeoxycholate (T DCA). Deconjugation of TDCA was demonstrated by a matte opaque granular appearance of the colonies. This phenotype was not observed for PR8240 (data not shown). 99 Problems with the assay arose when the screen was used for testing the bile salt hydrolase mutant strain, PRB240. This mutant demonstrated some growth defects, particularly on the glycine-conjugated bile acids (data not shown). The reduced growth presents a problem as the precipitation of deconjugated bile salts is dependent on acidification of the media surrounding the bacterial colonies (8), and this acidification depends on sufficient growth of the strain. In order to counteract this reduced growth, lower concentrations of bile acids were used. Under these conditions, the phenotypes for the wild-type strain were not as well defined, particularly the matte opaque granular colony phenotype noted on plates containing the taurine conjugated bile acids. Due to this discrepancy and to confirm that the various phenotypes shown in Figure 3.1 did represent deconjugation of the various bile acids, a different method, thin layer chromatography (TLC), was used for investigating the deconjugation activity of L. reuteri ATCC PTA 6475 and PRBZ40. For these studies, both L. reuteri ATCC PTA 6475 and PRB240 were grown in MRS plus 1 mM (approximately 0.05%) of each of the six major human bile acids separately. The strains were grown for 18 hours or 72 hours and tested for deconjugation activity by TLC. The TLC results confirmed that L. reuteri ATCC PTA 6475 is able to deconjugate all six major human bile acids by 18 hours; no activity was observed for PRB240, even after 72 hours of incubation (data not shown). Thus, L. reuteri ATCC PTA 6475 contains one bile salt hydrolase gene, 100 NT01LR0487, which is able to deconjugate both the taurine and glycine- conjugated forms of cholate, deoxycholate, and chenodeoxycholate. Disruption of NT01LR0487 leads to a growth defect in the presence of a synthetic human bile mixture. When L. reuteri ATCC PTA 6475 and PR8240 were grown in the presence of a synthetic human bile acid mixture (SHB), growth defects were observed for PRBZ40. The doubling time for L. reuteri ATCC PTA 6475 in MRS broth was 39 minutes; this time slowed to 48 i 1 minutes when the strain was grown in the presence of SHB. PRBZ40 had a doubling time of 42 i 4 minutes in the absence of bile; this time slowed to 63 i 1 minutes in the presence of bile. Bile inhibited the growth of both strains, although the inhibition observed for the bile-salt hydrolase mutant was more severe (a 23% increase in doubling time was observed for the wild-type strain compared to a 50% increase for PRB240). Growth in the presence of bile also decreased the final culture density that the strains were able to obtain. In the absence of bile, both strains reached a final 0.0.600 approximately equal to 4.7 after 24 hours, revealing that there is not inherent growth defect due to the disruption of the bile salt hydrolase gene, NT01LR0487. In the presence of bile, L. reuteri ATCC PTA 6475 reached a culture density of 1.91 i 0.24 (40% of that obtained without bile), whereas PR3240 reached a culture density of 0.8 i 0.01 (17% of that obtained without bile). 101 TLC confirmed that L. reuteri ATCC PTA 6475 was able to deconjugate a mixture of bile acids; the deconjugation products begin accumulating after 4 hours of growth (Figure 3.2), particularly for deoxycholate and/or chenodeoxycholate. No activity above the background level was observed for PRB240, even after 23 hours of incubation. 102 1. PRBZ40, 23 “Ours .\ 2. MRS (n99_ °°ntro|) :19 3'6475’1hOUr ,,‘H 4. 6475, 4 hams 5. 6475, 7 “Ours 6. 6475, 23 “Ours 7. CA (pos. control) V3 *Conjugated bile acids V003 IVOCI Figure 3.2. Deconjugated bile acids begin to accumulate after 4 hours in a culture of L. reuteri ATCC PTA 6475. Representative TLC plate showing the bile salt hydrolase activity of L. reuteri ATCC PTA 6475 against a synthetic “human” bile acid mixture (SHB). Samples were taken a 1, 4, 7, and 23 hours. Control solutions of cholate (CA) and deoxycholate (DCA) are included for comparison [deoxycholate and chenodeoxycholate (CDCA) migrate at the same rate in this solvent system]. The bile salt hydrolase mutant strain, PRB240, is shown to have no activity at the 23-hour time point. Controlling the pH of the growth medium alters the growth characteristics of the wild-type and mutant strains in the presence of a synthetic human bile mixture. Various reports in the literature have suggested that pH of the 103 environment may play an important role in the way that bacteria respond to bile acids. Previous work in the lab has demonstrated that L. reuteri is capable of lowering the pH of the growth medium from 6.5 down to 4.0. In order to determine whether this shift in pH played a role in the observed growth effects and/or deconjugation activity of L. reuteri ATCC PTA 6475 and PRB240, the cells were grown in a buffered version of MRS broth. Results from this set of growth curves, as well as the set described above are listed in Table 3.2. Table 3.2. Doubling times and culture densities for L. reuteri ATCC PTA 6475 and PRB240 in the presence and absence of human bile acids. Doubling time 0.0.500 (23- Strain Broth Bile (mirfl 24 hours) L. reuteri ATCC PTA 6475 MRS - 39 4.69 1 0.01 + 48 11 1.91 10.24 PRB240 - 42 1 4 4.71 1 0.01 + 6311 0801001 L. reuteri ATCC PTA MRS - 6475 acetate - 39 1 3 4.54 1 0.13 + 47 1 3 1.52 1 0.40 PRB240 - 48 1 9 4.73 10.08 + 66 + 11 0.70 1 0.07 L. reuteri Buffered ATCC PTA MRS - 6475 acetate - 37 1 5 2.41 1 0.09 + 44 1 4 2.31 10.22 PRB240 - 37 + 4 1.86 1 0.23 + 46 1 3 2.54 1 0.12 104 Interestingly, in the buffered medium, the differences between the wild-type strain and the bile salt hydrolase mutant regarding the lower final culture density and the slower doubling time are no longer observed. The lack of differences between the two strains are not due to a difference in deconjugation activity in this medium, as confirmed by TLC (Figure 3.3). These results demonstrate that pH does not affect the bile salt hydrolase activity of L. reuteri ATCC PTA 6475. The growth advantage provided by bile salt hydrolase activity, however, does appear to be pH-dependent. 105 1. MRS (neg. control) 2. Buff. MRS (neg. control) 3. PRBZ40, MRS 4. PRBZ40, Buff. MRS 5. 6475, MRS 6. 6475, Buff. MRS 7. CA (pos. control) 8. DCA (pos. control) O > *Conjugated bile acids V000 N00 Figure 3.3. Controlling the pH of the culture does not affect the bile salt hydrolase activity of L. reuteri ATCC PTA‘6475. Representative TLC plate showing the bile salt hydrolase activity of L. reuteri ATCC PTA 6475 against a synthetic “human" bile acid mixture (SHB) in MR8 and buffered MRS. Samples were taken at 7 hours. Control solutions of cholate (CA) and deoxycholate (DCA) are included for comparison [deoxycholate and chenodeoxycholate (CDCA) migrate at the same rate in this solvent system]. NO difference in activity is observed between cultures grown in MR8 and cultures grown in buffered MRS. Summary of the effects of various bile acids on growth of L. reuteri. Throughout the course of this research, various growth effects of particular bile acids were noted for L. reuteri ATCC PTA 6475 and PRBZ40. L. reuteri ATCC 55730 was also included in several of these studies for comparison. Although 106 these experiments were carried out using different concentrations of bile acids, and the strains were grown on MRS plates or in MRS broth, these trends remained consistent. These qualitative observations are summarized in Table 3.3. Table 3.3. Observed growth effects of individual bile acids in plates or in liquid medium on L. reuteri ATCC PTA 6475, PRBZ40, and L. reuteri ATCC 55730. Notable differences between ATCC PTA 6475 and PRB240 are highlighted in bold. N0 = not determined. *Concentrations of greater than 0.1% were not tested for the deconjugated bile acids due to solubility problems. L. reuteri L. reuteri ATCC PTA ATCC 6475 PRB240 55730 Cholate (CA) reduced reduced growth at growth at 0.1 % 0.1 % ND l Glycocholate (GCA) good growth good growth at 0.1% at 0.1% ND [Taurocholate (TCA) good growth good growth at 0.1% - at0.1% ND Deoxycholate (DCA) good growth good growth at 0.1% at 0.1% ND LGlycodeoxycholate (GDCA) good reduced reduced growth at growth at growth at 0.2% 0.05% 0.05% [ Taurodeoxycholate (TDCA) good growth good growth good growth at 0.5% and at 0.5% and at 0.5% and below below below 107 Table 3.3 (cont'd) Chenodeochholate QDCA) reduced reduced reduced growth at growth at growth at 0.05% 0.05% 0.05% Glycochenodeoxycholate GCDCA) severely reduced reduced reduced growth at growth at growth for 0.05% 0.05% 0.05% Taurochenodeoxycholate GCDCA) good growth good growth good growth at 0.5% at 0.5% at 0.5% In general, these growth studies suggest that the glycine-conjugated bile acids cause higher growth inhibition than the taurine-conjugated bile acids, and the deconjugated bile acids cause higher growth inhibition than either of the conjugated forms. PRBZ40, the bile salt hydrolase mutant, appears to be more sensitive to glycodeoxycholate and glycochenodeoxycholate, suggesting that the bile salt hydrolase activity may play a role in resistance to glyco-conjugated bile acids. 108 DISCUSSION The ability of bacteria to deconjugate bile acids is a trait that has been investigated both in an attempt to understand what benefit this enzyme may provide for the bacteria, as well as what effect the activity may have on the host. Several large-scale studies have demonstrated a strong correlation between the isolation of bacteria from the gastrointestinal tract and the ability of these bacteria to deconjugate bile acids (19, 27). This correlation suggests that this activity most likely confers some competitive advantage to strains, although the exact function has not been elucidated. Bile salt hydrolase activity of L. reuteri ATCC PTA 6475. These studies revealed that L. reuteri ATCC PTA 6475 contains one bile salt hydrolase enzyme, NT01LR0487. This enzyme is capable of deconjugating all six major bile acids found in human bile: taurocholate (TCA), taurodeoxycholate (TDCA), taurochenodeoxycholate (TCDCA), glycocholate (GCA), glycodeoxycholate (GDCA), and glycochenodeoxycholate (GCDCA). PRB240, the strain containing a disruption in NT01LR0487, was shown to have lost the ability to deconjugate these bile acids. The substrate specificity of bile salt hydrolase enzymes appears to be variable, as some enzymes exhibit specificity based on the steroid nucleus of the bile acid (cholate, deoxycholate, or chenodeoxycholate) and others exhibit specificity depending on the amino acid conjugate (taurine or glycine). The occurrence of bile salt hydrolase enzymes capable of 109 deconjugating all six human bile acids is unknown, as many studies have demonstrated deconjugation activities using only one or two bile acids. At least two other studies have demonstrated bile salt hydrolase enzymes capable of deconjugating the full range of bile acids (28, 30). Effect of bile salt hydrolase activity during growth in the presence of bile. Although there is a strong correlation connecting gastrointestinal strains of bacteria with the ability to deconjugate bile acids, in vitro and in vivo data demonstrating a direct relationship between the activity and persistence is conflicting. One of the main proposed functions of bile salt hydrolase activity is the detoxification of bile acids, although in vitro data regarding this is also variable. This study demonstrated that when the pH of the growth medium was not controlled (i.e., acidification due to metabolic products produced by the bacteria was allowed), bile salt hydrolase activity appeared to confer an advantage. This advantage was observed both as a decrease in doubling time and as an increase in the final culture density achieved in the presence of bile. This advantage was not present, however, when the pH of the medium was controlled (not allowed to drop below 6.5). In buffered medium, the doubling times and the final culture densities achieved by the wild-type strain and the bile- salt hydrolase mutant were virtually identical. The pH of the culture medium in the two situations was quite different, as in the unbuffered medium, the pH of the culture started at 6.5 and dropped down to 3.5 in as little as six hours (depending on the growth of the strain). In the buffered medium, the pH of the culture began 110 at 7.0 and only dropped down to 6.5, even after 24 hours of growth (data not shown). As most studies have demonstrated an acidic pH is necessary for optimum activity of bile salt hydrolase enzymes (7, 17, 28, 30), and indeed one study even observed full activity at a pH of 5.2 and a complete lack of activity at a pH of 6.8 (31 ), TLC was performed to determine what effect the varying pHs may have had on the bile salt hydrolase activity of L. reuteri ATCC PTA 6475. The TLC data demonstrated that there was no observable difference in the activity levels of the enzyme in the two media types (Figure 3.3). This data strongly suggests that the growth differences observed in the two types of media are due to an advantage of bile salt hydrolase activity in acidic medium. This phenomenon of differing effects of bile salt hydrolase activity at different pH values has been reported in the literature for other bacteria, primarily in regards to the glycine-conjugated bile acids, which are generally found to be more toxic than the taurine-conjugated forms. De Smet et al (10) demonstrated that at a low pH, glycodeoxycholate exhibited higher toxicity levels against strains with little to no bile salt hydrolase activity. Grill at al (16) also demonstrated that the toxic effect of glycodeoxycholate increased at pH values of less than 6.5 and showed that this effect was more pronounced for strains without bile salt hydrolase activity. Begley et al (4) also demonstrated that glycodeoxycholate exhibited higher levels of toxicity at pH 5.5 as compared to a pH of 6.5 for Listeria 111 monocytogenes and showed that this effect was even more pronounced in a bile salt hydrolase mutant. Interestingly, their data also demonstrated that the bile salt hydrolase mutant had a survival advantage over the wild-type strain when exposed to glycodeoxycholate at a pH above 5.5. These studies, along with the experiments performed herein, suggest that there is an. advantage to bile salt deconjugation at low pH levels. As the advantage seems to be observed somewhere between a pH of 5.5 and 6.5, and the pH of the small intestine ranges from 5.7 to 7.7 (25), it is difficult to surmise whether this activity contributes a significant advantage in vivo. Proposed model of bile salt toxicity through intracellular acidification. One proposed function for bacterial bile salt hydrolase activity is to detoxify the conjugated bile acids, which are proposed to inhibit cells through intracellular acidification in the same manner as organic acids. De Smet et al (10) outlined a model for this particular mechanism, including an explanation for the difference in toxicity observed for taurine versus glycine-conjugated bile acids. The authors propose that the protonated form of the conjugated bile acids is able to passively diffuse into the cell, where it promptly becomes deprotonated due to the alkaline intracellular environment. In a cell capable of bile salt hydrolase activity, the bile acid is then deconjugated, producing a compound with a higher pKa (around 5.0) whose conjugate base will pick up the extra proton before being transported outside of the cell, thus relieving the potential intracellular acidification. In a cell that is not capable of bile salt hydrolase activity, the cell must expend energy to 112 pump out the excess protons. The authors note that due to the lower pKa value of taurine-conjugated bile acids (pKa of around 0.9) versus glycine-conjugated bile acids (pKa of around 4.3), the concentration of protonated taurine- conjugated bile acids that could passively diffuse into the cell is very low at pH values achieved during bacterial growth. This reduced concentration is proposed to account for the reduced toxicity observed with these bile acids. The research presented here does not fully support this model. Although a higher level of growth inhibition is observed for the wild-type and mutant strains when grown in the presence of GDCA and GCDCA (compared to the taurine- conjugated equivalents), and the defect is more severe for the mutant than the wild-type strain, no such defect is observed when either strain is grown in the presence of GCA (Table 3.3). As all three glycine-conjugated bile acids have very similar pKa values, ranging from 4.27 to 4.34 (6), it is unlikely that the slight variations in the concentration of protonated bile acids would cause this difference in toxicity. Also, none of the taurine-conjugated bile acids exhibited an inhibitory effect on any of the L. reuteri strains tested. Despite the fact that taurine-conjugated bile acids are much stronger acids, and therefore the concentration of the protonated form that is proposed to enter the cell is very low at the pH values observed during the various culture conditions, deconjugation of all three forms of taurine-conjugated bile acids by L. reuteri ATCC PTA 6475 was observed. As most of the bile salt hydrolase enzymes described to date are intracellular enzymes (3, 30), the taurine-conjugated forms must enter the cell in 113 order to become deconjugated. The lack of toxicity observed for these bile acids against PRB240 (the mutant strain without any bile salt hydrolase activity) and for L. reuteri ATCC 55730 (which exhibits weak bile salt hydrolase activity specific to glycine-conjugated bile acids) suggests a different mechanism of toxicity besides passive diffusion leading to intracellular acidification. As many studies, including the research presented here, have demonstrated that the deconjugated bile acids are more inhibitory to bacteria than the conjugated bile acids (9, 14, 27, 29), the question of why bacteria would exhibit bile salt hydrolase activity still remains to be answered. One of the current theories specifies that the bacteria obtain some nutritional benefit from the activity, particularly through the use of the amino acid moieties (3). Nutritional benefit for L. reuteri appears to be an unlikely reason based on the results of this study, as both L. reuteri ATCC PTA 6475 and PRBZ40 exhibited equal doubling times and final culture densities during growth in the buffered medium, despite the presence and absence of bile acid deconjugation activity. Preliminary experiments performed to determine if the addition of taurine to the growth medium conferred a growth advantage also suggest that bile salt hydrolase activity does not confer a nutritional benefit (data not shown). This research demonstrates that bile salt hydrolase activity provides a growth advantage to L. reuteri ATCC PTA 6475 during growth at acidic pH values. The mechanism providing this growth advantage has not yet been elucidated. The 114 lack of an effect observed in buffered MRS medium suggests that the advantage is pH-dependent, although most of the data does not support the previous model of prevention of intracellular acidification due to passive diffusion of the conjugated bile acids into the cell. Further studies, including examination of the intracellular pH values of the cells under the various growth conditions, investigation of the potential role of a conjugated bile acid transporter, and closer r analysis of the specific rates of deconjugation for individual bile acids may lead to an understanding of the mechanism through which this advantage is provided to L. reuteri ATCC PTA 6475. Acknowledgements. The buffered MRS recipe was developed by Cathy Robinson. TLC was performed in the lab of Dr. Babak Borhan, with assistance from Daniel Whitehead. 115 10. REFERENCES Bateup, J. M., M. A. McConnell, H. F. Jenkinson, and G. W. Tannock. 1995. 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. 61:1147-1149. Begley, M., C. G. M. Gahan, and C. Hill. 2005. The interaction between bacteria and bile. FEMS Microbiol. Rev. 29:625-651. Begley, M., C. Hill, and C. G. M. Gahan. 2006. Bile salt hydrolase activity in probiotics. Appl. Environ. Microbiol. 72:1729-1738. Begley, M., R. D. Sleator, C. G. Gahan, and C. Hill. 2005. Contribution of three bile-associated loci, bsh, pva, and btlB, to gastrointestinal persistence and bile tolerance of Listeria monocytogenes. Infect. Immun. 73:894-904. Browning, J. D., and J. Sreenarasimhaiah. 2006. Gallstone disease, p. 1405-1412. In L. S. Friedman and L. J. Brandt (ed.), Sleisenger and Fordtran's Gastrointestinal and Liver Disease, 8th ed. Saunders, Philadelphia. Cantafora, A., A. Di Biase, M. Angelico, and M. Alvaro. 1987. Study of biologically relevant physical-chemical properties of bile salts by reverse- phase liquid chromatography. Chromatographia 24:277-281. Christiaens, H., R. J. Leer, P. H. Pouwels, and W. Verstraete. 1992. Cloning and expression of a conjugated bile acid hydrolase gene from Lactobacillus plantarum by using a direct plate assay. Appl. Environ. Microbiol. 58:3792-3798. Dashkevicz, M. P., and S. D. Feighner. 1989. Development of a differential medium for bile salt hydrolase-active Lactobacillus spp. Appl. Environ. Microbiol. 55:11-6. De Boever, P., and W. Verstraete. 1999. Bile salt deconjugation by Lactobacillus plantarum 80 and its implication for bacterial toxicity. J. Appl. Microbiol. 87:345-352. De Smet, I., L. Van Hoorde, M. Vande Woestyne, H. Christiaens, and W. Verstraete. 1995. Significance of bile salt hydrolytic activities of lactobacilli. J. Appl. Bacteriol. 79:292-301. 116 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. Delphino, M. V., M. I. Marchesini, S. M. Estein, D. J. Comerci, J. Cassataro, C. A. Fossati, and P. C. Baldi. 2007. A bile salt hydrolase of Bruce/la abortus contributes to the establishment of a successful infection through the oral route in mice. Infect. Immun. 75:299-305. Denou, E., R. D. Pridmore, B. Berger, J. M. Panoff, F. Arigoni, and H. Brussow. 2008. Identification of genes associated with the long-gut- persistence phenotype of the probiotic Lactobacillus johnsonii strain NCC533 using a combination of genomics and transcriptome analysis. J. Bacteriol. 190:3161-8. Dussurget, 0., D. Cabanes, P. Dehoux, M. Lecuit, C. Buchrieser, P. Glaser, and P. Cossart. 2002. Listeria monocytogenes bile salt hydrolase is a PrfA-regulated virulence factor involved in the intestinal and hepatic phases of listeriosis. Mol. Microbiol. 45:1095-1106. Elkins, C. A., and L. B. Mullis. 2004. Bile-mediated aminoglycoside sensitivity in Lactobacillus species likely results from increased membrane permeability attributable to cholic acid. Appl. Environ. Microbiol. 70:7200- 7209. Graham, 0. Y., and M. S. Osato. 2000. H. pylori in the pathogenesis of duodenal ulcer: interaction between duodenal acid load, bile, and H. pylori. Am. J. Gastroenterol. 95:87-91. Grill, J. P., S. Perrin, and F. Schneider. 2000. Bile salt toxicity to some bifidobacteria strains: role of conjugated bile salt hydrolase and pH. Can. J. Microbiol. 46:878-884. Hae-Keun, 0., J. Y. Lee, S. J. Lim, M. J. Kim, G. Kim, J. H. Kim, S. Hong, and D. Kang. 2008. Molecular cloning and characterization of a bile salt hydrolase from Lactobacillus acidophilus PF01. J. Microbiol. Biotechn. 18:449-456. Hofmann, A. F. 1979. The enterohepatic circulation of bile acids in man, p. 3-24. In G. Paumgartner (ed.), Bile Acids in Gastroenterology, vol. 6. Saunders, Philadelphia. Jones, B. V., M. Begley, C. Hill, C. G. Gahan, and J. R. Marchesi. 2008. Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proc. Natl. Acad. Sci. U. S. A. 105:13580-13585. Kurdi, P., H. Tanaka, H. W. Van Veen, K. Asano, F. Tomita, and A. Yokota. 2003. Cholic acid accumulation and its diminution by short-chain fatty acids in bifidobacteria. Microbiology 149:2031-7. 117 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. Law, J., G. Buist, A. Haandrikman, J. Kok, G. Venema, and K. Leenhouts. 1995. A system to generate chromosomal mutations in Lactococcus lactis which allows fast analysis of targeted genes. J. Bacteriol. 177:7011-8. Liong, M. T., and N. P. Shah. 2005. Bile salt deconjugation ability, bile salt hydrolase activity and cholesterol co-precipitation ability of lactobacilli strians. Int. Dairy J. 15:391-398. Maguin, E., P. Duwat, T. Hege, D. Ehrlich, and A. Gruss. 1992. New thermosensitive plasmid for gram-positive bacteria. J Bacteriol 174:5633- 8. Northfield, T. C., and I. McColl. 1973. Postprandial concentrations of free and conjugated bile acids down the length of the normal human small intestine. Gut 14:513-518. Ridlon, J. M., D. Kang, and P. B. Hylemon. 2006. Bile salt biotransformations by human intestinal bacteria. J. Lipid Res. 47:241-259. Russell, W. M., and T. R. Klaenhammer. 2001. Efficient system for directed integration into the Lactobacillus acidophilus and Lactobacillus gasseri chromosomes via homologous recombination. Appl. Environ. Microbiol. 67:4361-4. Tanaka, H., K. Doesburg, T. lwasaki, and I. Mierau. 1999. Screening of lactic acid bacteria for bile salt hydrolase activity. J. Dairy Sci. 82:2530- 2535. Tanaka, H., H. Hashiba, J. Kok, and I. Mierau. 2000. Bile salt hydrolase of Bifidobacterium Iongum-biochemical and genetic characterization. Appl. Environ. Microbiol. 66:2502-2512. Taranto, M. P., G. Perez-Martinez, and G. Font de Valdez. 2006. Effect of bile acid on the cell membrane functionality of lactic acid bacteria for oral administration. Res. Microbiol. 157:720-725. Taranto, M. P., F. Sesma, and G. F. de Valdez. 1999. Localization and primary characterization of bile salt hydrolase from Lactobacillus reuteri. Biotechnol. Lett. 21:935-938. Taranto, M. P., F. Sesma, A. Pesce de Ruiz Holgado, and G. F. de Valdez. 1997. Bile salts hydrolase plays a key role on cholesterol removal by Lactobacillus reuteri. Biotechnol. Lett. 19:845-847. 118 32. 33. Usman, and A. Hosono. 2000. Effect of administration of Lactobacillus gasseri on serum lipids and fecal steroids in hypercholesterolemic rats. J. Dairy Sci. 83:1705-1711. Walter, J., P. Chagnaud, G. W. Tannock, D. M. Loach, F. Dal Bello, H. F. Jenkinson, W. P. Hammes, and C. Hertel. 2005. A high-molecular- mass surface protein (Lsp) and methionine sulfoxide reductase B (MsrB) contribute to the ecological performance of Lactobacillus reuteri in the murine gut. Appl. Environ. Microbiol. 71:979-86. 119 CHAPTER 4 IDENTIFICATION OF A PUTATIVE NOVEL IMMUNOMODULATORY COMPOUND FROM LACTOBACILLUS REUTERI INTRODUCTION Probiotics are live microorganisms that confer health benefits to the host when administered in adequate amounts (6, 8). Many probiotics are now marketed to consumers and include organisms such as lactobacilli, streptococci, bifidobacteria, E. coli Nissle 1917, and the yeast Saccharomyces boulardii (12). In general, probiotic lactobacilli are considered safe for human consumption based on previous clinical trials, epidemiological studies, and historical usage in fermented foods (1, 23). However, the mechanisms by which probiotics promote good health and combat diseases are poorly understood. A better understanding of how probiotics influence the health of the host is critical to utilizing these organisms to their fullest potential. One emerging area of probiotic research is the ability of these bacteria to alter the immune system. Several probiotics secrete immunomodulins that modulate the host inflammatory response, but the bacterial products responsible for the effects on inflammation are still undefined (7, 17, 19). The inflammatory cytokine TNF, which is produced primarily by monocytes and macrophages, is a key 120 mediator of intestinal inflammation. lmmunoprobiotics, or probiotics producing key immunomodulatory factors, are potential therapies for various immune- mediated disorders such as Crohn’s disease (14). Indeed, select L. reuteri strains inhibit TNF production by monocytoid THP-1 cells and monocytes isolated from patients with Crohn’s disease (15, 19). Strains of L. reuteri capable of inhibiting TNF were able to reduce inflammation in a H. hepaticus-induced murine model of inflammatory bowel disease (19). The mechanism(s) behind this down regulation of inflammation are not understood. Bacterial species can be identified based on their fatty acid composition. In 1950, the first cyclopropane fatty acid (CFA) was identified as cis-11,12- methylene octadecanoic acid, or lactobacillic acid, in L. arabinosus (9). Additional CFAs have been identified in gram-negative and gram-positive bacteria, including dihydrosterculic acid (4). As shown in Figure 4.1, lactobacillic and dihydrosterculic acids are produced from the precursors, vaccenic acid and oleic acid respectively, by the enzyme cyclopropane fatty acid synthase (Cfa). Lactobacillic and dihydrosterculic acids are 19 carbon CFAs that differ only in the placement of the propane ring. During synthesis, Cfa adds a methylene carbon across the double bond forming a three-carbon ring using S-adenosylmethionine as a methyl donor. While our knowledge of how CFAs are produced has greatly increased in the past decade, the physiological role(s) of CFAs are poorly understood. CFAs may stabilize the cellular membrane, allowing bacteria to 121 survive exposure to adverse conditions such as extreme acid shocks and repeated freeze/thaw cycles (4). OH OH Oleic Acid Dihydrosterculic Acid V Cyclopropane Fatty Acid Synthase /OH—\ OH Vaccenic Acid 0 Lactobacillic Acid Figure 4.1. Synthesis of cyclopropane fatty acids. Cyclopropane fatty acid synthase converts oleic acid and vaccenic acid into dihydrosterculic acid and lactobacillic acid, respectively (4). Microbial fatty acids and lipids have been previously shown to have immunomodulatory properties. Lipid A is the active component of bacterial LPS, 3 known agonist of TLR2 and TLR4. The length and degree of saturation of the fatty acid chains of Lipid A are important determinants of LPS activity (18). Plasmodium falciparum produced a mixture of fatty acids that was more effective at inhibiting TNF production than any sole fatty acid tested (5). A Mycobacterium tuberculosis mutant unable to synthesize trans-cyclopropyl mycolic acids was 122 more virulent and stimulated murine TNF production by bone marrow-derived macrophages and RAW 264.7 macrophages more than the wild type strain (21 ). However, the effects of cyclopropane fatty acids produced by probiotic bacteria on the host inflammatory responses are poorly understood. This report demonstrates that synthesis of the specific CFA, lactobacillic acid, is restricted to TNF-inhibitory L. reuteri strains. The production of lactobacillic acid correlates with the emergence of the ability to inhibit TNF production. L. reuteri strains incapable of producing lactobacillic acid have an insertion of DNA at the 3’ end of the cfa gene, which may affect their ability to synthesize lactobacillic acid (Figure 4.7). We have constructed a L. reuten' cfa mutant and shown that it is unable to synthesize lactobacillic acid. The cfa mutant has partially lost the ability to inhibit TNF production in an LPS-activated macrophage model. Possible direct and indirect effects of lactobacillic acid on immunomodulation are discussed. MATERIALS AND METHODS Key reagents, bacterial strains and mammalian cell lines. All chemical reagents were obtained from Sigma-Aldrich (St. Louis, MO) unless otherwise stated. Polystyrene plates were obtained from Corning (Corning, NY). Polyvinylidene fluoride membrane filters of 0.22 pm pore size (Millipore, Bedford, MA) were used to yield L. reuteri-derived cell-free supernatants. All bacterial 123 strains and plasmids are described in Table 4.1. L. reuteri strains were cultured in deMan, Rogosa, Sharpe (Difco, Franklin Lakes, NJ) or LDMIIIG (supplemental material) media unless otherwise state. PRB173 was grown in the presence of 10 ug/mL erythromycin. Cis-vaccenic acid was ordered from MP Biomedicals (Ohio). For anaerobic culturing, an anaerobic chamber (1025 Anaerobic System, Forma Scientific, Waltham, MA) supplied with a mixture of 10% CO2, 10% H2, and 80% N2 was used. THP-1 cells (ATCC TlB-202) were maintained in RPMI 1640 supplemented with 10% fetal bovine serum (Invitrogen, Carlsbad, CA) at 37°C and 5% CO2. Table 4.1 — Bacterial strains and plasmids used in this study. Bacterial Strains and Plasmids Description Source Bacterial strains L. reuteriATCC Isolate from Peruvian mother’s Biogaia AB 55730 milk (Raleigh, NC) L. reuteri ATCC Isolate from Finnish mother’s Biogaia AB PTA 6475 milk (Raleigh, NC) L. reuteriATCC Oral isolate from Japanese Biogaia AB PTA 5289 female (Raleigh, NC) L. reuteri CF48-3A Fecal isolate from Peruvian Biogaia AB child (Raleigh, NC) L. reuteri PRB173 L. reuteri ATCC PTA 6475 This study pPBR173 inserted into cfa E. coli EC1000 chromosomal copy of the (13) pWV01 repA gene; Kanr Plasmids pVE6007 Cmr repA-positive temperature- (16) sensitive derivative of pWV01 pORl28 Emr repA-negative derivative of (13) pWV01 pPRB173 pORl28 + 239 bp insert from This study NT01LR1 143 124 Creation of PRB173 (L. reuteri ATCC PTA 6475 with a mutation in NT01LR1143). The following concentrations of antibiotics were used: 40 ug/mL kanamycin, 10 ug/mL chloramphenicol, 10 ug/mL erythromycin (L. reuten) or 400 ug/mL erythromycin (E. coli). The NT01LR1143 (cyclopropyl fatty acid synthase) mutant was created using the system developed by Russell and Klaenhammer (22) with modifications made for use in Lactobacillus reuteri (26). In short, 239 bp from NT01LR1143 from L. reuteri ATCC PTA 6475 was cloned into pORl28. This construct was then transformed into E. coli EC1000, and the transformed cells were grown in LB broth in the presence of erythromycin and kanamycin. pORl28 containing the NT01LR1143 insert was then extracted and transformed into L. reuteri ATCC PTA 6475 containing pVE6007. L. reuten' cells containing both plasmids were grown in MRS broth containing chloramphenicol and erythromycin at the permissive temperature of 35°C for 18 hours. The cultures were then shifted to the non-permissive temperature of 45°C and grown in the presence of erythromycin only. After several passages at 45°C to ensure loss of pVE6007, individual colonies were screened for integration of pORl28 at the desired location by PCR. Colonies were screened for the presence of both flanking regions (using one primer that anneals to pORl28 and one primer that anneals to the chromosome outside of the region cloned into pORl28) and the absence of a correctly-sized wild-type gene. 125 Samples for fatty acid methyl ester (FAME) analysis of overnight cultures (strain comparison and mutant analysis). Cultures for analysis were grown aerobically without shaking for 18 hours in 10 mL of MRS (Difco) at 37°C (L. reuteri CF48-3A was grown at 35°C by S. E. Jones). PRB173 was grown in the presence of 10 ug/mL erythromycin. Cultures were spun down at 4000 rpm for 10 minutes at room temperature. Supernatant was removed. Pellets were r resuspended in one ml of fresh MRS and spun at 13,400 rpm for 2 minutes. I Supernatant was removed and pellets were frozen at -80°C until analysis. i Experiments for strain comparisons were performed in quadruplicate. For mutant versus wild-type comparison, six wild-type samples were analyzed and four Ii mutant samples were analyzed. Samples for FAME analysis at various stages of growth. Cultures of L. reuten' ATCC PTA 6475 for analysis were grown in the presence of 2% oxygen with slow shaking in 110 mL of MRS (Difco) at 37°C. Culture density was measured at an optical density (0.0.) of 600 nm. 50 mL samples were collected from the cultures at mid-log (0.0. = 0.4); 10 mL samples were collected from the cultures at early stationary (0.0. = 3.1), late stationary (0.0. = 4.2), and after overnight incubation (approximately 18 hours). A representative growth curve is shown in Figure 4.2. After collection, samples were processed as described above, and pellets were frozen at -80°C until analysis. Experiments were performed in triplicate. 126 I 10 0.0.600 0.1 . 0.01 . T . fl 0 200 400 600 800 time (min) Flgure 4.2. A representative growth curve for L. reuten’ ATCC PTA 6475. Samples for fatty acid analysis (mid-log, early stationary, and late stationary phase) were collected at the time points represented by open circles. Samples for FAME analysis In dlfferent variations of MRS medla. Three variations of MRS media were used to determine the effects of various media components on the fatty acid composition of L. reuten‘ ATCC PTA 6475 and PRB173. Cultures were grown aerobically in 10 mL of MRS or modified MRS for 18 hours without shaking at 37°C. PRB173 was grown in the presence of 10 pg/mL erythromycin. Complete MRS (MRS A to distinguish it from the Difco MRS) was made from the following components: 10 g/L proteose peptone no. 3, 10 g/L beef extract, 5 g/L yeast extract, 20 g/L dextrose, 1 g/L Tween 80, 2 g/L ammonium citrate, dibasic, 5 g/L sodium acetate, 0.1 g/L magnesium sulfate, 0.05 g/L manganese sulfate, and 2 g/L dipotassium phosphate. Two variations of MRS media were also made. The first, MRS B, is MRS media made without 127 beef extract or Tween 80. The second, MRS C, is MRS media made without beef extract or Tween 80 with the addition of 0.01% cis-vaccenic acid (w/v). Samples were processed as listed above, and pellets were frozen at -80°C until analysis. Experiments were performed in triplicate. Fatty acid analysis. All fatty acid analysis was performed by Microbial lD Ir- (Newark, DE; mpzllwwwmicropjalidcom). Preparation of cell-free supernatants for immunomodulation studies. For '_ planktonic cells, 10 mL of LDMIIIG was inoculated at a starting 00600 = 0.1 (~7 x i 107 CF U) using 16-18 hr L. reuteri cultures. Bacteria were incubated for 24 hr at 35°C in anaerobic conditions. Serial dilutions were plated onto MRS medium to determine cell counts. Cells were pelleted (4000 x 9, RT, 10 minutes) and discarded. Supernatants were filter-sterilized. Aliquots were vacuum-dried and re-suspended to the original volume using RPMI. For biofilms, L. reuteri cultures (16-18 hrs of incubation) were diluted 1:100 in MRS and aliquots were placed into polystyrene 24-well plates. Plates were incubated anaerobically for 24 hr at 35°C. Supernatants and planktonic cells were removed by aspiration, and biofilms were washed with 50 mM sodium phosphate buffer (37°C, 100 rpm, 10 minutes). One mL of LDMIIIG was added to each well, and the plates were incubated for 2 hours at 35°C in anaerobic conditions. The supernatants were filtered through 0.22 gm filters, vacuum-dried and resuspended in RPMI to the 128 starting volume. Biofilms were removed by sonication (5 min, 20°C), and serial dilutions were plated to determine cell counts. TNF inhibition experiments. TNF assays were previously described (15). Briefly, cell-free supernatants from L. reuteri planktonic or biofilm cultures (5% v/v) or serial dilutions of lactobacillic acid (2% v/v) were added to human THP-1 cells (approximately 5 x 104 cells) activated with E. coli O127:B8 LPS (100 T ng/mL). After the addition of L. reuteri supernatants and LPS, plates were incubated at 37°C with 5% CO2 for 3.5 hours. Trypan blue (Invitrogen, Carlsbad, CA) was employed to ascertain cell viability. THP-1 cells were pelleted (1500 x i g, 5 min, 4°C), and quantitative ELISAs (R80 Systems, Minneapolis, MN) were used to determine the amount of TNF present in monocytoid cell supernatants. Statistical analyses for TNF bioassays and fatty acid profiles. For the TNF bioassays, a minimum of three biological replicates were performed and analyzed by ANOVA. Differences were considered statistically significant if p <0.05. Fatty acid profiles were determined a minimum of three times. All error bars in the figures represent standard deviations. RESULTS lmmunomodulatory activity of TNF inhibitory L. reuteri strains is produced upon entry into stationary phase. Some probiotic L. reuteri strains have the 129 ability to produce secreted factors that suppress lipopolysaccharide (LPS) induced TNF production in primary monocytes and macrophages (15). This activity is strain specific. We have identified two strains of L. reuteri (ATCC PTA 6475 and ATCC PTA 5289) that secrete one or more compounds that exhibit potent immunomodulatory activity against TNF production in activated macrophages. Secreted compounds from a third strain included in this study, L. reuteri ATCC PTA 4659, have also been shown to inhibit TNF production (3). T Two additional strains (L. reuteri ATCC 55730 and CF-48/3A) were incapable of reducing TNF production (11). The inhibitory effect on TNF production is found in culture supernatants of cells grown into stationary phase and is absent from t culture supernatants of exponentially growing L. reuteri. The goal of this study was to identify the immunomodulatory compounds produced upon entry into stationary phase using a combined genomics and genetic approach. The cyclopropyl fatty acid, lactobacillic acid, is specifically produced in immunomodulatory strains of L. reuteri. Fatty acids have been shown to play important roles in the regulation of the immune system. To identify potential membrane fatty acids that are produced specifically by immunomodulatory L. reuteri strains we compared the membrane fatty acid profiles of the three strains capable of inhibiting TNF production with the two strains that are incapable of downregulating TNF production. These five strains were grown to stationary phase and fatty acid methyl ester analysis was performed to determine the membrane fatty acid content. The results for the major fatty acids are shown in 130 Figure 4.3. Briefly, the immunomodulatory strains appeared to be very similar to one another; they consisted of approximately 41% palmitic acid (16:00), 11% Oleic acid (18:1 w9c), 11% vaccenic acid (18:1 (97c), 5% stearic acid (18:00), 18% dihydrosterculic acid (19:0 cyclo w9c), and 10% lactobacillic acid (19:0 cyclo (98c). The immunoneutral strains, L. reuteri ATCC 55730 and L. reuteri CF48- 3A, consisted of approximately 26% palmitic acid, 21% oleic acid, 17% vaccenic _- acid, 4% stearic acid, and 24% dihydrosterculic acid. Interestingly, the immunomodulatory strains contained a novel cyclopropyl fatty acid, lactobacillic L acid (phytomonic acid) that was absent from the immunoneutral strains. Thus .1 I t I lactobacillic acid is a possible candidate immunomodulin produced by L. reuteri strains ATCC PTA 6475, ATCC PTA 5289, and ATCC PTA 4659. A second cyclopropyl fatty acid, dihydrosterculic acid, was present in both immunoneutral and immunomodulatory strains in similar amounts. This indicated that immunoneutral strains were capable of producing fatty acids containing cyclopropane rings. 131 I Palmitic '3 Oleic Vaccenic l Stearic D Dihydrosterculic U Lactobacillic 45 40 35* 30 25 Percentage of total fatty acids 0 l, ”.4 ., ........_. . 1.1. .. ..- _. .... ._ .._..... L. “2.... LL . ATCC 55730 CF48-3A ATCC PTA ATCC PTA ATCC PTA 5289 4659 6475 Figure 4.3 - TNF-inhibitory strains produce lactobacillic acid. Cultures of five strains of L. reuteri were grown in MRS broth for 18 hours at 37° C. Fatty acid profiles were determined by gas chromatography. ATCC 55730 and CF48- 3A (TNF non-inhibitory strains) do not produce the CFA, lactobacillic acid. In contrast TNF-inhibitory strains (ATCC PTA 6475, ATCC PTA 5289, and ATCC PTA 4659) produce lactobacillic acid. Experiments were performed in triplicate; error bars represent standard deviation. * Lactobacillic acid. The appearance of lactobacillic acid correlates with the appearance of the immunomodulatory activity. Previous work has demonstrated that the TNF inhibitory activity is only found in cell-free supernatant from L. reuten' cultures that have entered stationary phase. In order to investigate whether there was a correlation between the presence of lactobacillic acid in the cell membranes and the presence of the immunomodulatory compound in the cell-free supernatants, 132 fatty acid profiles were compared from L. reuteri ATCC PTA 6475 cultures at various stages of growth. According to the representative growth curve in Figure 4.2, samples for fatty acid analysis were collected from cultures at mid-log phase (0.0.600 = 0.4), early stationary phase (0.0.500 = 3.0), and late stationary phase (0.0.600 = 4.1). Samples were also collected after overnight incubation of cultures (approximately 18 hours). Lactobacillic acid was not present in cultures in mid-log phase or early stationary phases of growth (Figure 4.4). Cultures at late stationary phase contained approximately 9.6% lactobacillic acid; overnight cultures contained approximately 10.3%. Dihydrosterculic acid, the other cyclopropyl fatty acid present in cultures of L. reuteri, was found at concentrations of approximately 20% for all four time points tested (Figure 4.4). Thus, the presence of lactobacillic acid in cultures of L. reuteri ATCC PTA 6475 correlates with the appearance of the TNF inhibitory compound(s) in cell-free supernatant from the immunomodulatory strains. 133 lpalmitic loleic Dvaccenic Ddihydrosterculic Elactobacillic Percentage of fatty acids * Ir- ‘ Mid-log Early stationary Late stationary Overnight Figure 4.4. Lactobacillic acid appears in late stationary phase cultures of L. reuteri ATCC PTA 6475. Cultures of L. reuteri ATCC PTA 6475 were grown in MRS broth at 37°C. Samples were collected for FAME analysis at different stages of growth. Experiments were performed in triplicate; error bars represent standard deviation. *Lactobacillic acid. The production of lactobacillic acid is due to the activity of NT01LR1143, a cyclopropyl fatty acid synthase. Previous work in E. coli has shown that the biosynthesis of bacterial cyclopropyl fatty acids is due to one enzyme, cyclopropyl fatty acid synthase. This cyclopropyl fatty acid synthase (Cfa) is responsible for the conversion of oleic acid into dihydrosterculic acid and the conversion of vaccenic acid into lactobacillic acid (Figure 4.1). A search of the L. reuteri ATCC PTA 6475 genome revealed a single gene, NT01LR1143, with significant similarity to known cyclopropyl fatty acid synthases. A disruption in 134 the gene was created by insertion of pORl28 as described in the Materials and Methods section. The resulting strain was titled PRB173. In order to confirm the effect of the disruption of NT01LR1143, cultures of L. reuteri ATCC PTA 6475 and PRBI73 were grown overnight in MRS or MRS plus 10 ug/mL erythromycin. Samples were collected, and fatty acid analysis was performed. The results, shown in Figure 4.5, confirmed that the mutant strain, PRB173, was devoid of I... lactobacillic acid and contained a minor amount of dihydrosterculic acid (1.6% 9 compared to 17.8% in wild-type). The mutant strain also contained a higher j) amount of oleic acid (52.5%) compared to the wild-type strain (10.7%). ; :a?‘ , it 135 I ATCC PTA 6475 PRB173 .8601 850— '1‘ 540 E 930 n- u— I 320 O) 310 C L3 g 0 Ir Figure 4.5. Disruption of NT01LR1143 (CFA synthase) eliminates production of lactobacillic acid by L. reuteri ATCC PTA 6475. L. reuteri ATCC PTA 6475 and PRB173 were grown overnight in MRS broth at 37° C. Samples were collected for fatty acid analysis. Experiments were performed in triplicate; error bars represent standard deviation. Removal of Tween 80 from the growth medium Increases the amount of lactobacillic acid found In L. reuteri ATCC PTA 6475. By disrupting the cyclopropyl fatty acid synthase gene (NT01LR1 143) of L. reuteri ATCC PTA 6475, a strain was created that does not contain lactobacillic acid (PRB173). This strain will serve as an important control for future experiments directed at determining the possible role of lactobacillic acid in suppression of TNF by stimulated macrophages. A strain with an increased concentration of lactobacillic 136 acid would also be useful for these studies. To this aim, attempts were made to increase the percentage of lactobacillic acid in L. reuteri ATCC PTA 6475 cultures through alteration of specific media components. L. reuteri cultures are commonly grown in MRS broth, a rich undefined media, in the laboratory. One component of MRS broth is Tween 80, which contains approximately 70% Oleic acid with a balance of palmitic acid, stearic acid, and linoleic acid. Dihydrosterculic acid, the only other cyclopropyl fatty acid present in cultures of L. reuteri is made from oleic acid, and previous work in other lactobacilli has demonstrated that removal of oleic acid from the medium can increase the production of lactobacillic acid (10). In addition, the effect of adding 0.01% cis-vaccenic acid (the precursor for lactobacillic acid) in the absence of Tween 80 was also determined. L. reuteri ATCC PTA 6475 was grown in one of three variations of MRS broth for 18 hours, and samples were collected for fatty acid analysis. The three variations of broth used were: MRS A (complete MRS made from the various components), MRS B (MRS media without Tween 80 or beef extract), and MRS C (MRS B plus 0.01% cis-vaccenic acid). A summary of the results is shown in Figure 4.6. Briefly, in the absence of Tween 80 (MRS B), the lactobacillic acid concentration increased from approximately 8.7% of the total fatty acids to 29.9%, whereas the amount of dihydrosterculic acid decreased from 15.9% to 0% in the absence of its precursor. The addition of cis-vaccenic acid (MRS C) lead to a small increase in the lactobacillic acid, making it a total of 33.5% of the fatty acids found in the samples. Thus, growth of L. reuteri ATCC 137 PTA 6475 in either MRS B or MRS C would provide another sample for future studies aimed at investigating the role of lactobacillic acid in immunomodulation. PRB173, the CFA synthase mutant, was confirmed to be completely devoid of lactobacillic acid in MRS B (data not shown). 138 IMRSA IMRSB DMRsc 50 45- 40- g .6 35— (0 B30— .12 To - E 25 ‘1' “6 _ a, 20 9 E15— 9 8310‘ 5_ 04 . '0 “s <9 Q‘b Flgure 4.6. Lactobacillic acld production by L. reuteri ATCC PTA 6475 can be increased by removal of Tween 80 from the growth medium. Overnight cultures of L. reuteri ATCC PTA 6475 were grown in MRS A (complete MRS broth), MRS B (MRS broth with Tween 80 and beef extract removed), or MRS 0 (MRS B plus 0.01% cis-vaccenic acid); samples were collected for fatty acid analysis. Experiments were performed in triplicate; error bars represent standard deviation. 139 The cyclopropyl fatty acid synthase gene is truncated in strains of L. reuteri incapable of down-regulating TNF production. Membrane cyclopropane fatty acids (CFA) have been shown to increase as cells enter stationary phase. CFA formation is performed by the enzyme cyclopropane fatty acid synthase that is encoded by the cfa gene. We therefore scanned the genomes of L. reuteri PTA ATCC 6475 and ATCC 55730 and identified a single cfa gene in each genome. Interestingly, the reading frame of the cfa gene was truncated at the 3’ end by the insertion of a DNA element in the L. reuteri ATCC 55730 but not the ATCC PTA 6475 strains (Figure 4.7). The insertion in L. reuten' ATCC 55730 results in a C-terminal deletion of 12 amino acids in the Cfa protein. To determine if this insertion correlated with the loss of immunomodulatory activity we PCR amplified the cfa gene and flanking sequences. The insertion was identified in both immunoneutral strains tested but was absent in both strains capable of suppressing TNF. Thus the absence of the insertion correlated with the ability to reduce TNF production. 140 ATCC PTA 6475 and ATCC PTA 5289 LR2652 RibBA "_‘ HP l Insertion Figure 4.7. An insertion in the CFA synthase gene of L. reuteri ATCC 55730 and CF48-3A may prevent production of lactobacillic acid. PCR analysis revealed an insertion in the region of the genome containing the cfa gene for both immunomodulatory strains. This insertion results in the truncation of the cfa gene. Small arrows represent the primers used for amplification of the region surrounding the cfa gene. Conditioned medium from a mutant defective in cyclopropyl fatty acid production is impaired in inhibiting TNF production. To address what possible role lactobacillic acid production has on the ability to inhibit TNF production we compared the effects of conditioned media from wild-type cells and the cfa mutant (PRB173) on the ability of activated monocytes to produce TNF. Human derived THP-1 monocytes were activated by the addition of LPS and the production of TNF was monitored by quantitative ELISA. Cell-free 141 supernatants from L. reuteri deficient in cyclopropane fatty acid synthase (PBR173) partially lost the ability to suppress TNF when compared to the immunomodulatory effects of wild type ATCC PTA 6475 in both planktonic cultures and biofilms (Figure 4.8). For planktonic cultures, TNF production was efficiently suppressed by cell-free F” supernatant derived from L. reuteri ATCC PTA 6475 compared to the LDMIIIG '_ medium control. The cfa mutant stain PRB173 was deficient in its ability to I! reduce TNF production compared to the wild-type strain. TNF production was approximately 2-fold higher in bioassays with PBR173 compared to bioassays E’ with wild type L. reuteri ATCC PTA 6475 (239 pg/mL and 134 pg/mL, respectively) (p<0.001) (Figure 4.8 A). In addition, serial dilutions of probiotic- derived supernatants were tested. At a dilution of 1:20, wild-type supernatants still inhibited TNF by 45% compared to the media control. In contrast, at a 1:20 dilution, PRB173 supernatants had completely lost their TNF-inhibitory ability (data not shown). We also analyzed the production of immunomodulatory activity from biofilms and found similar trends with respect to TNF modulation. When supernatants from biofilm cultures were tested, ATCC PTA 6475 and PRB173 inhibited TNF production by 60% and 21%, respectively, compared to the medium alone (Figure 4.8 B). In the presence of supernatants derived from the Cfa-deficient strain, the quantities of human TNF produced by LPS-activated THP-1 cells were approximately 2-fold higher than quantities observed in the wild type strain (560 pg/mL and 282 pg/mL, respectively) (p < 0.01). Planktonic and 142 biofilm cultures of the immunoneutral L. reuteri ATCC 55730 did not suppress TNF when compared to the media control (Fig. 4.8). In addition, mutations in NT01LR0396 (multi-drug resistance transporter in the major facilitator superfamily) and NT01LR1106 (putative esterase) had no effect on the ability of L. reuteri ATCC PTA 6475 to suppress TNF, demonstrating the mutagenesis system had no effect (data not shown). These results demonstrate that mutants defective in lactobacillic acid production are unable to fully suppress TNF suppression. 143 1 200 1000 800 TNF (pg/mL) O) O O .b O O 200 Media D < 0.001 ATCC PTA 6475 PBR173 ATCC 55730 ' B 1 000 800 O) O O TNF (pg/mL) A O O 200 Media D < 0.01 ATCC PTA 6475 PBR173 ATCC 55730 144 Figure 4.8 - L. reuteri deficient in Cfa had diminished ability to suppress TNF. Cell-free supernatants from L. reuteri cultured as planktonic cells or biofilms were tested for the ability to inhibit LPS-activated THP-1 cells from producing TNF. When cultured as planktonic cells (A) or biofilms (B), PBR173, which does not produce lactobacillic acid, had diminished ability to suppress TNF production compared to wild-type cells. DISCUSSION The ability of probiotics to affect the immune system of the host is likely to be an important mechanism for how these types of bacteria benefit human health. Several gastrointestinal disorders, including Crohn’s disease, are partly caused by increased inflammation in the intestine, and therapies that target pro- D} inflammatory cytokines such as TNF have been effective in providing symptomatic relief is some cases. Thus candidate probiotics that can downregulate pro-inflammatory cytokines in the intestinal tract offer a means of y localized delivery of an immunomodulatory agent. Several studies have identified potent immunomodulatory activities expressed by candidate probiotic bacteria, however in most cases the bacterial factors mediating these activities are not understood. In this study we have identified the cyclopropane fatty acid lactobacillic acid as a candidate immunomodulin involved in the inhibition of TNF produced by activated macrophages. Several lines of evidence support that L. reuteri strains that produce lactobacillic acid have increased capabilities of inhibiting TNF. First, the presence of lactobacillic acid was only found in strains capable of down- regulating TNF production. Second, the appearance of lactobacillic acid in stationary phase correlates with the appearance of immunomodulatory activity. Third, mutation of cfa, which eliminates the production of lactobacillic acid, 145 results in a L. reuteri strain with reduced capacity to inhibit TNF production from both planktonic cultures and biofilms. These results indicate that lactobacillic acid is either an immunomodulatory compound itself or loss of lactobacillic acid indirectly effects the production of another immunomodulin. We have had lactobacillic acid synthesized and have not detected TNF inhibition by lactobacillic acid at non-cytotoxic concentrations (data not shown). This either indicates that lactobacillic acid does not directly play a role in suppression of TNF expression or that we have not identified the proper conditions for delivering lactobacillic acid to macrophages. This could be due to either biological or technical reasons. We found that lactobacillic acid was very insoluble as a free fatty acid and therefore it is unclear that we were successful in delivering lactobacillic acid properly to the macrophages. In other situations fatty acids delivered to cells in phospholipids are able to mediate effects not observed with free fatty acids (7). We are currently attempting to use petroleum ether extracted lipids from the immunomodulatory L. reuteri strains to determine if we can reconstitute immunomodulatory activity. The effects of cyclopropanation in lipids on the immune response has recently been studied in Mycobacterium tuberculosis. The disruption of cyclopropyl ring formation in mycolic acids has a profound effect on how this pathogen interacts with the immune system. Deletion of the cmaZ gene, which encodes a cyclopropane synthase enzyme which adds trans-cyclopropyl rings to mycolic 146 II. [Ila-"'2. “Ill . .1 .- '31; «.31 acids, produces a hypervirulent strain in a mouse model of tuberculosis. Induction of pro-inflammatory cytokines in murine-derived macrophages was more pronounced with the mutant strain and this could be mimicked with phenyl ether extracted lipids. This supports the claim that trans-cyclopropanation of mycolic acids attenuates the host response to M. tuberculosis (21). Interestingly, deletion of the pcaA gene, which encodes another cyclopropyl mycolic acid synthase in M. tuberculosis and is responsible for the synthesis of cis-cyclopropyl rings in mycolic acids, had the opposite effect on virulence and in vitro inflammatory responses (20). This work indicates that cyclopropane ring fatty acids can influence pathogenicity and the host immune response. Whether or not lactobacillic acid has a similar direct effect remains to be determined. Although we currently favor a direct effect of lactobacillic acid on the inflammatory response of LPS-stimulated macrophages, it is equally possible that the observed effects of deletion of Cfa are indirect. We have identified 36 genes that are significantly reduced in expression when cfa is disrupted, each of these are candidates for being involved in immunomodulation (Dephine Saulnier and Jim Versalovic, personal communication). The most interesting gene from this group is a putative cyclase(NT01LR1833) that is found only in some strains of L. reuteri but not other lactobacilli. Interestingly, L. reuteri ATCC 55730 does not contain this gene, again correlating the absence of this gene with a lack of immunomodulatory activity. The function of this cyclase is unknown. NT01LR1833 is located in a putative operon with a cobalamin-independent 147 methionine synthase and near an enzyme that catalyzes the final step in menaquinone (vitamin K) biosynthesis. Vitamin K, which is an essential vitamin for humans, has been linked to the regulation of the immune response. We are currently investigating whether or not this putative cyclase has any role in immunomodulation. Further work is needed to determine if lactobacillic acid plays a direct or indirect role in modulation of the host immune response. The creation of a mutant strain of L. reuteri ATCC PTA 6475, PRB173, that is incapable of producing lactobacillic acid, as well as the identification of growth conditions that triple the lactobacillic acid content of the cell membranes will aid in these pursuits. Acknowledgements. This project was carried out in collaboration with researchers at Baylor College of Medicine. Sara E. Jones grew the overnight cultures of L. reuteri CF48-3A (a second immunoneutral strain) for fatty acid analysis and performed the TNF inhibition experiments (including growth of bacterial and THP—1 cells, preparation of cell-free conditioned medium, and TNF assays). Delphine Saulnier identified‘the truncation in the cfa synthase gene of the immunoneutral L. reuteri strains. The author would also like to acknowledge Daniel Whitehead for production of Figure 4.1. 148 10. REFERENCES Adams, M. R. 1999. Safety of industrial lactic acid bacteria. J. Biotechnol. 68:171-178. Altschul, S. F., W. Gish, W. Miller, E. W. Myers, and D. J. Lipman. 1990. Basic local alignment search tool. J. Mol. Biol. 215:403-410. Connolly, E. 2008. Selection and use of lactic acid bacteria for reducing I] inflammation in mammals. USA. 3 Cronan, J. E., Jr. 2002. Phospholipid modifications in bacteria. Curr. Opin. Microbiol. 5:202-205. y _- Debierre-Grockiego, F., L. Schofield, N. Azzouz, J. Schmidt, C. Santos de Macedo, M. A. Ferguson, and R. T. Schwarz. 2006. Fatty acids from Plasmodium falciparum down-regulate the toxic activity of malaria glycosylphosphatidylinositols. Infect. Immun. 74:5487-5496. FAO. 2001. Health and nutritional properties ofprobiotics in food including powder milk with live lactic acid bacteria: report of a joint FAONVHO expert consultation on evaluation of health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria. Food and Agriculture Organization. Frick, J. S., K. Schenk, M. Quitadamo, F. Kahl, M. Koberle, E. Bohn, M. Aepfelbacher, and l. B. Autenrieth. 2007. Lactobacillus fermentum attenuates the proinflammatory effect of Yersinia enterocolitica on human epithelial cells. Inflamm. Bowel Dis. 13:83-90. Fuller, R. 1989. Probiotics in man and animals. J. Appl. Bacteriol. 66:365- 378. Hofmann, K., and R. A. Lucas. 1950. The chemical nature of a unique fatty acid. J. Am. Chem. Soc. 72:4328-4329. Johnsson, T., P. Nikkila, L. Toivonen, H. Rosenqvist, and S. Laakso. 1995. Cellular Fatty Acid Profiles of Lactobacillus and Lactococcus Strains in Relation to the Oleic Acid Content of the Cultivation Medium. Appl. Environ. Microbiol. 61 :4497-4499. 149 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. Jones, S. E., and J. Versalovic. 2009. Probiotic Lactobacillus reuteri biofilms produce antimicrobial and anti-inflammatory factors. BMC Microbiol. 9:35. Jonkers, D., and R. Stockbrugger. 2007. Probiotics in gastrointestinal and liver diseases. Aliment. Pharmacol. Ther. 26 Suppl. 2:133-148. Law, J., G. Buist, A. Haandrikman, J. Kok, G. Venema, and K. Leenhouts. 1995. A system to generate chromosomal mutations in Lactococcus lactis which allows fast analysis of targeted genes. J. Bacteriol. 177:7011-8. Lin, Y., and J. Versalovic. 2006. lmmunoregulation by probiotics. AgroFOOD Industry Hitech. 17:56-59. Lin, Y. P., C. H. Thibodeaux, J. A. Pefia, G. D. Ferry, and J. Versalovic. 2008. Probiotic Lactobacillus reuteri suppress proinflammatory cytokines via c-Jun. lnflamm. Bowel Dis. 14:1068-1083. Maguin, E., P. Duwat, T. Hege, D. Ehrlich, and A. Gruss. 1992. New thermosensitive plasmid for gram-positive bacteria. J Bacteriol 174:5633- 8. Menard, S., C. Candalh, J. C. Bambou, K. Terpend, N. Cerf- Bensussan, and M. Heyman. 2004. Lactic acid bacteria secrete metabolites retaining anti-inflammatory properties after intestinal transport. Gut 53:821-828. Netea, M. G., M. van Deuren, B. J. Kullberg, J. M. Cavaillon, and J. W. Van der Meer. 2002. Does the shape of lipid A determine the interaction of LPS with Toll-like receptors? Trends Immunol. 23:135-139. Pefia, J. A., A. B. Rogers, Z. Ge, V. Ng, S. Y. Li, J. G. Fox, and J. Versalovic. 2005. Probiotic Lactobacillus spp. diminish Helicobacter hepaticus-induced inflammatory bowel disease in interleukin-10-deficient mice. Infect. Immun. 73:912-920. Rao, V., N. Fujiwara, S. A. Porcelli, and M. S. Glickman. 2005. Mycobacterium tuberculosis controls host innate immune activation through cyclopropane modification of a glycolipid effector molecule. J. Exp. Med. 201:535-543. Rao, V., F. Gao, B. Chen, W. R. Jacobs, Jr., and M. S. Glickman. 2006. Trans-cyclopropanation of mycolic acids on trehalose dimycolate suppresses Mycobacterium tuberculosis -induced inflammation and virulence. J. Clin. Invest. 116:1660-1667. 150 17.7 III-n2. ‘ ‘5'“: IT'.‘ .5" . A‘I". 22. 23. 24. 25. 26. 27. Russell, W. M., and T. R. Klaenhammer. 2001. Efficient system for directed integration into the Lactobacillus acidophilus and Lactobacillus gasseri chromosomes via homologous recombination. Appl Environ Microbiol 67:4361-4. Saxelin, M., N. H. Chuang, B. Chassy, H. Rautelin, P. H. Makela, S. Salminen, and S. L. Gorbach. 1996. Lactobacilli and bacteremia in southern Finland, 1989-1992. Clin. Infect. Dis. 22:564-566. Smyth, G. K., Y. H. Yang, and T. Speed. 2003. Statistical issues in cDNA microarray data analysis. Methods Mol. Biol. 224:111—136. Wall, T., K. Bath, R. A. Britton, H. Jonsson, J. Versalovic, and S. Roos. 2007. The early response to acid shock in Lactobacillus reuteri involves the CIpL chaperone and a putative cell wall-altering esterase. Appl. Environ. Microbiol. 73:3924-3935. Walter, J., P. Chagnaud, G. W. Tannock, D. M. Loach, F. Dal Bello, H. F. Jenkinson, W. P. Hammes, and C. Hertel. 2005. A high-molecular- mass surface protein (Lsp) and methionine sulfoxide reductase B (MsrB) contribute to the ecological performance of Lactobacillus reuteri in the murine gut. Appl Environ Microbiol 71 :979-86. Yang, Y. H., S. Dudoit, P. Luu, D. M. Lin, V. Peng, J. Ngai, and T. P. Speed. 2002. Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res. 30:e15. 151 CHAPTER 5 SUMMARY AND SIGNIFICANCE Although research in the field of probiotics has increased exponentially over the past twenty years, the mechanisms by which these organisms exert a beneficial effect on the host are not well understood. Much research has focused on in vitro studies investigating survival of stresses that the bacteria will be exposed to in the gastrointestinal tract, the ability to inhibit pathogens, and the ability to modulate the host immune response. In vivo studies are investigating a wide range of disorders including bacterial infections, allergies, and gastrointestinal disorders. Although various strains of L. reuteri have demonstrated effectiveness in various in vitro and in vivo studies as discussed in Chapter 1, the mechanisms by which this effectiveness is achieved has remained elusive. For example, there appears to be wide ranging differences between the physiology and effects of particular strains within the species, and very little has been determined regarding the actual interactions between L. reuteri and the host. Strain specificities. Differences between strains of L. reuteri need to be thoroughly investigated. Initial studies have demonstrated that there are differences in the stress resistance, production of antimicrobial compounds, immune response modulation, and effectiveness in animal trials. For example, one of the common characteristics of L. reuten' that is often attributed for the 152 ability of the species to cause beneficial effects is the production of the wide spectrum antimicrobial compound, reuterin. Research has shown that at least one strain, L. reuteri RC-14 does not produce reuterin, nor does it contain the necessary enzyme needed to convert glycerol into reuterin (4). Thus, any probiotic effects observed with this strain are not due to reuterin production, although this observation does not rule out reuterin’s importance for other beneficial effects. These investigations have identified some of the factors involved in the survival of L. reuteri in the host during exposure to bile in the small intestine, as well identified a putative novel immunomodulatory compound, lactobacillic acid. These studies have also highlighted some of the strain specificities that may play important roles in interactions with the host. Althoughall L. reuteri strains included in this work were of human origin, differences were observed in the mechanisms used for survival in the presence of bile. L. reuteri ATCC PTA 6475 appears to obtain a benefit from bile salt hydrolase activity during growth in the presence of bile. Preliminary research suggests that this strain’s bile salt hydrolase enzyme is active against a wider range of bile acids and acts more quickly than the enzyme from L. reuteri ATCC 55730. Interestingly, several of the genes identified as playing an important role in the survival and growth of L. reuten' ATCC 55730 in the presence of bile, the putative esterase (Lr1516) and the multidrug resistance protein (Lr1584), did not appear to have overlapping roles in L. reuteri ATCC PTA 6475 (data not shown). Whether or not this 153 difference is due to the bile salt hydrolase activity of L. reuteri ATCC PTA 6475 has yet to be determined. Another important strain difference is represented by the ability of some of the strains to suppress TNF, while others do not have an effect on TNF production by macrophages as described in Chapter 4. This difference suggests that some strains of L. reuten' may be effective in suppressing host inflammatory responses, while other strains may have no effect or may even exacerbate inflammation. Fatty acid analysis of these strains revealed that all five strains investigated had the ability to synthesize cyclopropyl fatty acids (all strains produced dihydrosterculic acid, a 19-carbon cyclopropyl fatty acid); however, only the immunomodulatory strains contained a second 19-carbon cyclopropyl fatty acid, lactobacillic acid. Although the prevalence of lactobacillic acid among the various strains, as well as the occurrence of lactobacillic acid at particular stages of growth, suggest a role for the compound in TNF suppression, it is possible that other strain differences are responsible. Bacterial-host interactions. While strain specificity is important to investigate, ultimately the main area lacking in probiotic research, despite much effort from the field, is an understanding of the mechanisms through which these microorganisms, like L. reuteri, are able to cause their beneficial effects. It is unlikely that one common mechanism of action will be identified, thus careful research investigating cautiously chosen strains in a range of host genetic backgrounds should be carried out. If one strain does not alleviate a particular 154 disease, another strain, with different traits, should be considered. Unfortunately, the situation could be even more complicated, as at least one study involving L. reuteri has demonstrated that the health status of the host can alter the response to probiotic administration (3). Ouwehand et al (9) also demonstrated that the health status of the host tissue could alter adherence levels of probiotic bacteria. L. reuteri ING1 exhibited better adherence to colonic tissue isolated from IBD patients than healthy patients, as well as better adherence to mucus from diseased tissue versus healthy tissue. Things such as inflammatory markers, other bacteria, or mucus composition could alter this interaction (9). One area to investigate potentially novel mechanisms of action or bacterial-host interactions would be genes that are up-regulated during stresses found in the host gastrointestinal system that do not appear to play a role in bacterial survival of this stress. The putative matrix metalloprotease discussed in Appendix C is an example of such a gene. Microarray analysis revealed that Ir1291 was up- regulated in L. reuteri ATCC 55730 during bile shock exposure, yet mutational analysis revealed that this gene does not appear to play a role in either survival or growth in the presence of bile. As bile is known to act as a signal for some pathogens to up-regulate virulence factors (5), it is possible that it may also act as a signal for commensals or probiotic strains to up-regulate factors involved in interaction with the host. The sequence similarity between lr1291 and eukaryotic matrix metalloproteases, along with the important roles of matrix 155 metalloproteases within the gastrointestinal tract (7, 8, 14), make this a particularly intriguing gene to investigate. Despite all of the unknown aspects of probiotics, there is substantial clinical and experimental evidence of their beneficial effects. L. reuteri appears to have great promise as probiotic species, exhibiting many of the in vitro characteristics classically considered to be “necessary” for a strain to cause beneficial effects, such as being of host origin, surviving stresses present in the gastrointestinal tract, and producing antimicrobial compounds. Importantly there are also multiple L. reuteri strains exhibiting beneficial effects in clinical trials, including RC-14’s effects on urogenital health (1, 2, 6) and ATCC 55730’s effects on general health and diarrhea in children (10-13). A better understanding of how L. reuteri responds to situations within the host and possibly modifies the responses of the host may lead to further applications for this species. 156 REFERENCES Anukam, K., E. Osazuwa, l. Ahonkhai, M. Ngwu, G. Osemene, A. W. Bruce, and G. Reid. 2006. Augmentation of antimicrobial metronidazole therapy of bacterial vaginosis with oral probiotic Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14: randomized, double-blind, placebo controlled trial. Microbes Infect. 8:1450-1454. Anukam, K. C., E. Osazuwa, G. I. Osemene, F. Ehigiagbe, A. W. Bruce, and G. Reid. 2006. Clinical study comparing probiotic Lactobacillus GR-1 and RC-14 with metronidazole vaginal gel to treat symptomatic bacterial vaginosis. Microbes Infect. 8:2772-2776. Baroja, M. L., P. V. Kirjavainen, S. Hekmat, and G. Reid. 2007. Anti- inflammatory effects of probiotic yogurt in inflammatory bowel disease patients. Clin. Exp. Immunol. 149:470-479. Cadieux, P., A. Wind, P. Sommer, L. Schaefer, K. Crowley, R. A. Britton, and G. Reid. 2008. Evaluation of reuterin production in urogenital probiotic Lactobacillus reuteri RC-14. Appl. Environ. Microbiol. 74:4645- 4649. Gunn, J. S. 2000. Mechanisms of bacterial reSistance and response to bile. Microbes Infect. 2:907-13. Martinez, R. C., S. A. Franceschini, M. C. Patta, S. M. Quintana, R. C. Candido, J. C. Ferreira, E. C. De Martinis, and G. Reid. 2009. Improved treatment of vulvovaginal candidiasis with fluconazole plus probiotic Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14. Lett. Appl. Microbiol. 48:269-274. Medina, C., and M. W. Radomski. 2006. Role of matrix metalloproteinases in intestinal inflammation. J. Pharmacol. Exp. Ther. 318:933-938. Naito, Y., and T. Yoshikawa. 2005. Role of matrix metalloproteinases in inflammatory bowel disease. Mol. Aspects Med. 26:379-90. Ouwehand, A. C., S. Salminen, P. J. Roberts, J. Ovaska, and E. Salminen. 2003. Disease-dependent adhesion of lactic acid bacteria to the human intestinal mucosa. Clin. Diagn. Lab. Immunol. 10:643-646. 157 10. 11. 12. 13. 14. Shornikova, A. V., I. A. Casas, E. lsolauri, H. Mykkanen, and T. Vesikari. 1997. Lactobacillus reuteri as a therapeutic agent in acute diarrhea in young children. J. Pediatr. Gastroenterol. Nutr. 24:399-404. Shornikova, A. V., l. A. Casas, H. Mykkanen, E. Salo, and T. Vesikari. 1997. Bacteriotherapy with Lactobacillus reuteri in rotavirus gastroenteritis. Pediatr. Infect. Dis. J. 16:1103-1107. Tubelius, P., V. Stan, and A. Zachrisson. 2005. Increasing work-place healthiness with the probiotic Lactobacillus reuteri: a randomised, double- blind placebo-controlled study. Environ. Health 4:25. Weizman, Z., G. Asli, and A. Alsheikh. 2005. Effect of a probiotic infant r? formula on infections in child care centers: comparison of two probiotic agents. Pediatrics 115:5-9. Wilson, C. L., A. J. Ouellette, D. P. Satchell, T. Ayabe, Y. S. Lopez- _ , . Boado, J. L. Stratman, S. J. Hultgren, L. M. Matrisian, and W. C. E] * Parks. 1999. Regulation of intestinal q-defensin activation by the metalloproteinase matrilysin in innate host defense. Science 286:113-117. 158 APPENDIX A MICROARRAY ANALYSIS OF LACTOBACILLUS REUTERI ATCC 55730 DURING MID-LOG AND EARLY STATIONARY PHASES OF GROWTH PURPOSE The purpose of this investigation was to compare the gene expression profiles of a commonly used probiotic strain, L. reuteri ATCC 55730, during mid-log and early stationary phases of growth. Collaborators at Baylor College of Medicine identified that certain strains of L. reuteri produce compounds that are able to suppress TNF production by activated macrophages (as described in Chapter 4). This activity is found in cell-free conditioned medium isolated from cells at the stationary phase of growth. The microarray studies described herein were performed in an attempt to identify potential targets to investigate as the possible immunomodulatory compounds. L. reuteri ATCC 55730 is not one of the strains that exhibits the TNF-suppressive activity, yet it was the only sequenced strain at the time of the experiments. MATERIALS AND METHODS Bacterial strains and growth conditions. L. reuteri ATCC 55370 was grown in MRS broth (BD Difco) at 37°C under microaerobic conditions (2% 02, and 5% 159 w: “Mi-"j 002, balanced with N2) with slow shaking, or grown on MRS agar plates incubated anaerobically using the GasPack EZ Anaerobe Container system (BD Difco) at 37°C unless otherwise specified. RNA isolation. L. reuten' ATCC 55730 from an 18-hour old culture was inoculated into 40 ml of MRS broth to a starting 0.0.500 approximately equal to 0.03. Cultures were grown to late stationary phase. For each of the five biological replicate experiments, 5 ml samples were collected at mid-log phase (O.D.600 approximately equal to 0.3) and early stationary phase (0.0.600 approximately equal to 2.4) and immediately mixed with an equal portion of ice- cold methanol to halt transcription. Cells were pelleted by centrifugation at 4000 rpm for 10 minutes and washed with STE buffer (6.7% sucrose, 50 mM Tris-Cl, pH 8.0, 1 mM EDTA). The washed cells were resuspended in 0.25 UhltS/lll mutanolysin (Sigma) dissolved in STE buffer and incubated at 37°C for 20 minutes. RNA was then isolated from lysed cells using the Qiagen RNeasy Kit with optional on column DNase treatment according to manufacturer’s instructions. Microarray experiments. Microarray experiments were performed as previously described (5). In short, 60-mer oligonucleotides for 1864 open reading frames were synthesized for the L. reuteri ATCC 55730 genome (1 ); 15 open reading frames from L. reuteri DSM20016 encoding extracellular proteins were also incuded. Various E. coli genes with no known similarity to the L. reuteri genome 160 were included as controls. The synthesized oligonucleotideswere spotted onto Corning UltraGAPS-ll slides; with each gene represented once on the microarray, while the control spots were spotted eight times, once in each subgrid. All work, from the design of the oligonucleotides to the array construction were performed at the Research Technology Support Facility at Michigan State University, East Lansing, MI, USA. RNA isolation, labeling, and hybridization were performed as previously described (3, 4). (Information regarding the microarray platform can be found at NCBls Gene Expression Omnibus (GEO, h_ttp://www.ncbi.nlm.nih.gov/geo/) under GEO platform number GPL6366). Five biological replicates, along with technical replication through dye-swapping, were performed for the microarray experiments. In other words, each RNA sample was subjected to two hybridizations and values used for subsequent analysis were averaged. Microarray data was analyzed using iterative outlier analysis with three iterations as previously described (2, 3). RESULTS Microarray experiments were performed to investigate the differences in the gene expression profiles of L. reuteri ATCC 55730 cultures at mid-log and early stationary phases of growth. The points of samples collection are shown in Figure A.1, which shows a representative growth curve for the strain. Overall, 85 161 genes were found to be significantly over-expressed as the cultures entered stationary phase, and 41 genes were found to be significantly under-expressed based on the iterative outlier analysis. A list of genes with significantly different expression patterns can be found in Tables A1 to A2. 10 - 1 _ O .8 Q o 0.1 ~ 0.01 T l l. l 0 100 200 300 400 time (min) Figure A.1. A representative growth curve of L. reuten' ATCC 55730. Samples for microarray analysis (mid-log and early stationary phase) were taken at the time point represented by the open circles. 162 Table A.1. Genes significantly over-expressed by L. reuteri ATCC 55730 during early stationary phase. 163 Accession Gene a Fold- name no. Functional classification change_ Energy production and conversion lr0904 NS Malate dehydrogenase (EC 1.1.1.37) 5.1 Ir1475 NS Malolactic enzyme 2.6 Ir1744 NS L-2-hydroxyisocaproate dehydrogenase 4.9 bifunctional protein: alcohol dehydrogenase; acetaldehyde lr1777 EF421920 dehydrogenase 3.5 lr1793 EF421923 1,3-propanediol dehydrogenase 3.2 lr1866 D0233710 PduW putative acetate kinase 7.6 COG1454 EutG Alcohol lr1867 D0233711 dehydrogenase class IV 11.6 PduP putative NAD-dependent Ir1868 D0233712 aldehyde dehydrogenase 10.3 PduA propanediol utilization protein: Ir1882 D0233726 putative microcompartment protein 4.9 Ir1919 NS Malate dehydrogenase (EC 1.1.1.37) 2.9 Amino acid transport and metabolism lr0054 NS Homoserine kinase (EC 2.7.1.39) 2.9 Ir0124 NS Amino acid permease 4.6 lr0125 EF537904 aminotransferase 6.1 lr0213 NS Aminotransferase 6.3 Cystathionine beta-Iyase/cystathionine lr0324 EF421 866 gamma-synthase 6.0 lr0529 NS Arginine/ornithine antiporter 5.6 lr0530 NS Arginine/ornithine antiporter 4.0 Ir0560 NS Cysteine synthase (EC 4.2.99.8) 8.5 Transcriptional regulator, GntR family / TYROSINE AMINOTRANSFERASE lr0621 NS (EC 2.6.1.5) 2.7 4-aminobutyrate aminotransferase (EC Ir0650 NS 2.6.1.19) 3.8 Ir0701 NS Aspartokinase (EC 2.7.2.4) 4.2 lr1019 DQ857773 ArcC, Carbamate kinase 4.7 COGOO78 ArgF Ornithine Ir1020 D0233707 carbamoyltransferase partial CDS 7.2 lr1113 EF534269 Threonine synthase 2.7 Table A.1 (cont’d) Aspartate aminotransferase (EC lr1162 NS 2.6.1.1) 5.1 Ir1258 EF421900 Xaa-Pro aminopeptidase, PepP 3.8 Ir1517 D0233695 COGZZ35 ArcA Arginine deiminase 6.6 COG0624 ArgE Acetylornithine deacetylase/Succinyl-diaminopimelate desuccinylase and related deacylases Ir1731 DQ233708 partial CDS 7.9 Ir1784 D0857803 Peptidase U34 3.1 PduU putative ethanolamine utilisation Ir1865 DQ233709 protein 9.8 Ir1881 DQ233725 PduB propanediol utilization protein 8.0 Carbamoylphosphate synthase small Ir2088 D0857861 subunit 2.6 Nucleotide transport and metabolism Dihydroorotase and related cyclic Ir0302 D0857868 amidohydrolases 4.6 Ir0521 DQ857762 Dihydroorotate dehydrogenase 3.0 Carbohydrate transport and metabolism lr1525 EF534272 Arabinose efflux permease 3.1 CobC, AIpha-ribazole-5‘-phosphate Ir1985 DQ857854 phosphatase 3.2 Coenzyme transport and metabolism 2-dehydropantoate 2-reductase (EC lr0969 NS 1.1.1.169) 2.8 Lipid transport and metabolism lr0149 NS Putative lipase/esterase 2.7 (acyl-carrier-protein) S- lr1009 EF421883 malonyltransferase, FabD 24.4 3-oxoacyI-[acyl-carrier-proteinj Ir1011 EF421885 synthase Ill, FabH 5.9 3-hydroxymyristoyl/3-hydroxydecanoyl- Ir1013 EF421887 (acyl carrier protein) dehydratase, FabA 23.7 EnoyI-[acyl-carrier protein] reductase Ir1279 EF421902 (NADH), Fabl 19.1 AcetyI-CoA carboxylase alpha subunit, lr1280 EF421903 AccA 16.3 AcetyI-CoA carboxylase beta subunit, lr1281 EF421904 Ach 14.3 Ir1282 EF421905 Biotin carboxylase, AccC 20.7 3-hydroxymyristoyl/3-hydroxydecanoyl- Ir1283 EF421906 (acyl carrier protein), FabA 16.1 164 lf’fflLfln .- u— .- j Table A.1 (cont’d) lr1284 EF421907 Biotin carboxyl carrier protein, AccB 18.1 3-oxoacyI-(acyl-carrier—protein) |r1286 EF421908 synthase, FabB 21.4 3-oxoacyI-(acyl-carrier—protein) Ir1287 EF421909 reductase, FabG 19.5 Transcription lr1012 EF421886 Transcriptional regulator, MarR family 20.7 Ir1310 EF421911 hypothetical protein , 4.2 COG1438 ArgR Arginine repressor lr1518 DQ233704 partial CDS 5.2 lr1573 NS Leucine-responsive regulatory protein 3.2 Replication, recombination and repair ATPase related to the helicase subunit lr0044 EF421958 of the Hollidayjunction resolvase 4.7 Posttranslational modification, protein turnover, chaperones lr0107 NS Thioredoxin 6.1 Inorganic ion transport and metabolism COG1464 NIpA ABC-type metal ion transport system periplasmic lr0629 DQO74834 component/surface antigen 3.6 COG1464 NlpA ABC-type metal ion transport system periplasmic Ir1151 DQ074882 component/surface antigen 3.7 lr2077 NS Copper-exporting ATPase (EC 3.6.3.4) 2.6 Secondary metabolites biosynthesis, transport and catabolism 2-oxo—hepta-3-ene-1,7-dioic acid lr1745 NS hydratase 3.3 lr1873 DQ233717 PduL propanediol utilisation protein 9.6 PduE propanediol dehydratase small Ir1878 DQ233722 subunit 4.6 PduC propanediol dehydratase large lr1880 DQ233724 subunit 8.7 PduN propanediol utilization protein: Ir1871 DQ233715 putative microcompartment protein 7.2 PduJ propanediol utilization protein: lr1874 DQ233718 putative microcompartment protein 4.5 PduK propanediol utilization protein: |r1875 DQ233719 putative microcompartment protein 1 1.3 General function prediction only |r0079 EF471975 D-lactate dehydrogenase 6.1 lr0272 EF537899 flavodoxin 5.6 Ir0313 EF421952 HAD-superfamily hydrolase 2.6 165 Table A.1 (cont’d) Ir11 15 NS NAD-dependent oxidoreductase 3.4 Ir1163 NS Nitrilase (EC 3.5.5.1) 4.0 lr1834 NS Nitrilase (EC 3.5.5.1) 4.8 lr1869 DQ233713 PduObis propanediol utilization protein 9.2 Signal transduction mechanisms lr0628 DQZ33673 LuxS protein 3.7 COG0834 HisJ ABC-type amino acid transport/signal transduction systems Ir0793 DQ074847 periplasmic component/domain 4.2 Ir2123 D0857864 Protein-tyrosine phosphatase 8.7 Unknown function Ir1010 EF421884 Acyl carrier protein, AcpP 12.4 Uncharacterized conserved protein, |r1047 EF421890 DegV 3.7 lr1319 DQ074896 COG4716 Myosin-crossreactive antigen 3.7 Ir1515 DQ074905 Unknown extracellular protein 2.9 PduO putative propanediol utilization lr1870 DQ233714 B12 related protein 3.8 Ir1872 DQ233716 PduM propanediol utilization protein 10.9 PduH putative diol dehydratase Ir1876 DQ233720 reactivation protein 6.1 PduG putative diol dehydratase lr1877 DQ233721 reactivation protein 7.2 PduD propanediol dehydratase medium lr1879 DQ233723 subunit 8.1 Ir2078 NS unknown protein 3.9 a GenBank accesion numbers are provided (NS = not submitted) b Genes were classified based on COG domains found in the protein sequence through a search of the JGI Integrated Microbial Genomes database. 166 Table A2. Genes significantly under-expressed by L. reuteri ATCC 55730 during early stationary phase. Accession Gene a Fold- name no. Functional classification change Energy production and conversion [Citrate [pro-3S]-lyase] ligase (EC lr0599 DQZ33700 6.2.1 .22) 5.0 NAD-dependent malic enzyme (EC Ir0600 DQ240820 1.1.1.38) 5.6 COG3051 CitF Citrate lyase alpha Ir1418 DQ233691 subunit 4.3 Amino acid transport and metabolism spermidine/putrescine ABC Ir0137 EF537897 transporter, permease protein 5.3 Glutamine transport ATP-binding lr1197 NS protein gan 5.1 COGO834 HisJ ABC-type amino acid transport/signal transduction systems periplasmic component/domain COG0765 HisM ABC-type amino acid transport system permease lr1 198 DQ074887 component 7.0 Ir1508 NS Gluconate permease 2.9 Histidine transport ATP-binding Ir1664 NS protein hisP 3.2 amino acid permease-associated N886:1 NS region 3.0 Nucleotide transport and metabolism lr0092 EF421961 Deoxyribose-phosphate aldolase 3.2 Ir0115 DQ219946 COG1428 Deoxynucleoside kinase 4.1 Pyrimidine-nucleoside phosphorylase Ir0674 NS (EC 2.4.2.2) 3.3 Carbohydrate transport and metabolism lr0101 EF421948 Phosphopentomutase (EC 5.4.2.7) 2.9 lr0160 EF547651 Sugar kinase, ribokinase family 13.7 lr0590 NS Deoxyribose transporter 3.9 lr0675 00857878 Phosphopentomutase 2.9 COG2301 CitE Citrate lyase beta lr1419 DQ233692 subunit 5.8 NS359:2 NS Ribose transport protein 12.6 167 Table A2 (cont'd) Coenzyme transport and metabolism COG1767 CitG Triphosphoribosyl- Ir1417 DQZ33690 dephosgho-COA synthetase 5.4 Lipid transport and metabolism N8207z1 NS Transcription regulator 5.8 Translation, ribosomal structure and biogenesis Ir0050 NS LSU ribosomal protein L14P 2.9 C060594 RnpA RNase P protein lr0251 DQ219950 component 3.2 tRNA (Guanine-N(1)-)- lr1551 NS methyltransferase (EC 2.1.1.31) 3.5 Transcription lr0647 NS Catabolite control protein A 3.2 Replication, recombination and repair lr1237 NS DNA gyrase subunit A (EC 5.99.1.3) 3.6 lr1238 NS DNA gyrase subunit A (EC 5.99.1.3) 3.4 lr1239 NS DNA gyrase subunit B (EC 5.99.1.3) 3.6 COG1195 RecF Recombinational lr1240 DQ219962 DNA repair ATPase 4.1 Cell motility putative type II secretion system NS376:1 NS protein 7.0 General function prediction only |r0517 NS Zinc protease (EC 3.4.99.-) 3.2 COG0596 MhpC Predicted hydrolases or acyltransferases Ir1416 DQZ33689 (alpha/beta hydrolase superfamily) 3.2 |r1666 NS putative zinc-dependent peptidase 4.2 N8337z1 NS Conserved hypothetical protein 4.3 Signal transduction mechanisms Stress response membrane GTPase Ir0279 EF421950 TypA 3.0 COG0834 HisJ ABC-type amino acid transport/signal transduction systems Ir1665 DQO74920 periplasmic component/domain 3.5 Intracellular trafficking secretion, and vesicular transport COG0706 Preprotein translocase |r0252 DQO74813 subunit YidC 3.0 Unknown function Ir0646 DQ233675 Hypothetical membrane protein 3.0 lr0982 D0857874 Hypothetical protein 3.4 168 Table A2 (cont’d) Ir1434 DQ074902 Unknown extracellular protein 3.1 lr1487 DQ074904 Unknown extracellular protein 4.1 NS104:1 NS unknown protein 3.4 a GenBank accesion numbers are provided (NS = not submitted) b Genes were classified based on COG domains found in the protein sequence through a search of the JGI Integrated Microbial Genomes database. The microarray experiments described were performed to identify potential immunomodulatory compounds based on gene expression changes as L. reuteri transitions from mid-log to early stationary phases of growth. Although L. reuteri ATCC 55730 is not one of the strains capable of producing compounds that suppress TNF production, it was the only strain for which sequence information and therefore, microarrays, were available at the time of the study. The microarray studies revealed two main pathways that were up-regulated as the bacteria transitioned from mid-log to early stationary phase, propanediol utilization and fatty acid biosynthesis. The interest in fatty acid biosynthesis led to the identification of fatty acid differences between the immunomodulatory and immunoneutral strains as described in Chapter 4. This data is not included in Chapter 4 because preliminary experiments carried out by the lab at Baylor College of Medicine using microarrays designed for one of the immunomodulatory strains, L. reuteri ATCC PTA 6475, did not confirm similar gene expression changes for this strain. This may be due to genuine differences in gene expression between the two strains or due to the collection of 169 samples at different time points (and possibly different stages of growth) between the two labs. 170 REFERENCES Bath, K., S. Roos, T. Wall, and H. Jonsson. 2005. The cell surface of Lactobacillus reuteri ATCC 55730 highlighted by identification of 126 extracellular proteins from the genome sequence. FEMS Microbiol. Lett. 253:75-82. Britton, R. A., P. Eichenberger, J. E. Gonzalez-Pastor, P. Fawcett, R. Monson, R. Losick, and A. D. Grossman. 2002. Genome-wide analysis of the stationary-phase sigma factor (sigma-H) regulon of Bacillus subtilis. J. Bacteriol. 184:4881-90. Uicker, W. C., L. Schaefer, and R. A. Britton. 2006. The essential GTPase RbgA (quF) is required for 508 ribosome assembly in Bacillus subtilis. Mol. Microbiol. 59:528-40. Wall, T., K. Bath, R. A. Britton, H. Jonsson, J. Versalovic, and S. Roos. 2007. The early response to acid shock in Lactobacillus reuten' involves the CIpL chaperone and a putative cell wall-altering esterase. Appl. Environ. Microbiol. 73:3924-3935. Whitehead, K., J. Versalovic, S. Roos, and R. A. Britton. 2008. Genomic and genetic characterization of the bile stress response of probiotic Lactobacillus reuteri ATCC 55730. Appl. Environ. Microbiol. 74:1812-1819. 171 APPENDIX B IDENTIFICATION OF A PUTATIVE BILE-INDUCIBLE TRANSCRIPTIONAL ELEMENT FROM LACTOBACILLUS REUTERI PURPOSE Bile has been demonstrated to serve as a signal for particular bacteria regarding their location in the gastrointestinal tract. For example, certain pathogens, 3, ._ including Vibrio, Shigella, Salmonella, and Campy/obacter, are known to alter ’ expression of virulence factors such as toxin production, motility, and type III secretion systems in response to bile (2). Various bile acids have also been shown to affect the germination of Clostridium difficilespores (3, 4). As there is known to be overlap in the interaction of pathogenic and commensal bacteria with the host, It is possible that bile also acts as a signal for probiotic bacteria. Using the microarray analysis of the bile shock response of L. reuteri ATCC 55730 (5), the purpose of this study was to use a bioinformatic approach to identify a bile-inducible transcriptional element associated with promoters induced by bile. 172 RESULTS The upstream regions (300 bp) of all genes over-expressed during the bile shock response of L. reuteri ATCC 55730 were analyzed for over-represented motifs using the freely-available internet program MEME (1). As it is unlikely that all of the genes over-expressed during bile shock will be regulated by the same element, multiple MEME runs were performed comparing the upstream regions of smaller subsets of genes. One statistically significant 13 base pair motif was identified when the upstream regions of 13 genes over-expressed during bile shock were compared. The upstream regions included in this particular run were from the following genes: lr0359, lr0540, lr0685, lr0783, lr0922, Ir1067, Ir1139, Ir1351, lr1468, Ir1684, lr1786, lr1864, and Ir1937. The sequence of the motif at each of these locations, as well as the consensus sequence is shown in Figure 8.1. 173 Gene number Motif sequence |r0359 CAATTGCGTCATC Ir1067 CAATTGCTTGATC |r1786 CAATTGCATAATC Ir0685 CAATTGGGTTATC lr1351 CAGTTGCCTTATC Ir0922 CATTTGCTTCATC lr1468 GAATTGCATCATC lr0783 CAATTGGGTGAAC lr1139 CAATTGCATAATT Ir1937 CAATTCCTTCATG lr1684 CCGTCGCGTTATC |r0540 CAATTGCCAAATG lr1864 TAAATGCGTTAAC [ConsensuslclAIAlrlrlclcGTCAITIcI A T T A Figure 8.1. The sequences of the 13 occurrences of the possible bile-inducible transcriptional element in L. reuteri ATCC 55730 identified by MEME. In collaboration with C. Titus Brown at Michigan State University, genome-wide analysis of this putative bile-inducible transcriptional element was performed. Dr. Brown identified that this particular 13 base pair sequence was found a total of 70 times throughout the L. reuteri ATCC 55730 genome and 54 times throughout the genome of another probiotic strain, L. reuteri ATCC PTA 6475. If this were a random sequence element it would be expected to be found only 11 times in a genome with the size (approximately 2.3 mega-bases) and GC content of L. reuteri ATCC 55730. 174 Further analysis revealed that of the 70 occurrences of this particular 13 base pair element in L. reuteri ATCC 55730, 66 of them are within the coding regions of other genes. It was also demonstrated that none of the additional 57 elements (beyond the original 13 identified) are located upstream of genes that are significantly over-expressed or under-expressed during the bile shock response of this strain. FUTURE DIRECTIONS Current work in the lab is focused on developing a reporter gene system for use in L. reuteri. This system will ultimately allow us to identify whether this putative 13 base pair transcriptional element is a bile-inducible promoter. A bile-inducible promoter could have great potential for application in the probiotic industry, as it would allow for targeted expression of a gene product in the host small intestine. 175 REFERENCES Bailey, T. L., and C. Elkan. 1994. Presented at the Proceedings of the Second International Conference on Intelligent Systems for Molecular Biology, Menlo Park, California. Gunn, J. S. 2000. Mechanisms of bacterial resistance and response to bile. Microbes and Infection 2:907-913. Sorg, J. A., and A. L. Sonenshein. 2008. Bile salts and glycine as cogerminants for Clostridium difficile spores. J. Bacteriol. 190:2505-2512. Sorg, J. A., and A. L. Sonenshein. 2009. Chenodeoxycholate is an inhibitor of Clostridium difficile spore germination. J. Bacteriol. 191:1115- 1 1 17. Whitehead, K., J. Versalovic, S. Roos, and R. A. Britton. 2008. Genomic and genetic characterization of the bile stress response of probiotic Lactobacillus reuteri ATCC 55730. Appl. Environ. Microbiol. 74:1812-1819. 176 APPENDIX C INVESTIGATION OF A PUTATIVE MATRIX METALLOPROTEASE FROM LAC TOBA CILLUS REUTERI PURPOSE The purpose of this study was to investigate the role of a putative metalloprotease identified in Lactobacillus reuteri. This gene, lr1291, was identified as being significantly over-expressed during the bile shock response of L. reuteri ATCC 55730. Mutational analysis did not reveal a role for this gene in survival or growth in the presence of bile (14). Sequence analysis revealed that ' the protein has significant similarity to eukaryotic matrix metalloproteases (MMPs), a class of proteins known to be involved in a wide range of important functions such as recruitment of immune cells, cytokine and chemokine activation, and degradation of the extracellular matrix. As very little is currently understood regarding how probiotic bacteria interact with the host to cause their beneficial effects, the presence of a gene with significant similarity to eukaryotic proteins known to play important roles in the gastrointestinal tract is of interest. Note: Although the gene was originally identified in L. reuteri ATCC 55730 (Ir1291), the homolog found in L. reuteri ATCC PTA 6475 (NT01LR0595) was used for all bioinformatic and over-expression studies discussed herein. There is 177 96% similarity between Lr1291 and NT01LR0595 at the amino acid level, and 93% similarity at the nucleic acid level; both proteins are referred to as Lr1291 for simplicity. BIOINFORMATICS A BLAST of the protein sequence of Lr1291 against the non-redundant protein sequences in GenBank revealed that the closest matches were peptidases in the M10A or M12B family (including matrixins and adamalysins), putative metalloproteinases, or zinc-dependent proteases in either Lactobacillus spp. or Streptococcus spp (1 ). Lactic acid bacteria with similar proteins included primarily host-associated strains such as lactobacilli commonly used as probiotics, including L. reuteri, L. acidophilus, L. gasseri, L. johnsonii, and L. plantarum, as well as pathogenic streptococci including Streptococcus pneumoniae and S. mutans. The significant hits also included several human matrix metalloproteases (MMPs) that aligned with the second half of Lr1291. A search of the conserved domain database (5) revealed that the last 130 amino acids of Lr1291 contains cd04268 (ZnMc_MMP_Iike subfamily). This group includes matrix metalloproteases, serralysins, and astacin-Iike family of proteases. To further investigate the sequence of Lr1291, the protein sequence was compared to the MEROPS database (10). This search revealed that Lr1291 fits 178 into the M10 family, although it was classified along with other unassigned peptidases. The active site for the M10 family is HExxHxxgxxH, including the histidine residues that act as zinc ligands and the catalytic glutamate. This family shares the same active site as several other families of metalloproteases (M12, M43, and M57). The M10 family peptidases are classified as “metzincins” because of the conserved methionine residue found C-terminal to the active site. The M10 family peptidases often are also found to function outside of the cell. In line with this, Lr1291 is predicted to be secreted, due to the presence of an N- terminal signal peptide without any discernable cell envelope anchor (2). Figure 0.1 shows the protein sequence of Lr1291with several characteristics of interest marked. >NT01 LR0595 VNEINFIFRLLKRIFIlGLLVGGGWLYFNDARVQA II TANQTAWNVRDRIAKLIGRDDTNSNDNSNLHLNDANNGSSKNEQGPTTEQQT STQTSIPSTGRWATNQATVYVNTNNAQLDAATNTAIQNWNQTGAFTFKPVNNQ SKADIVVTTMNRSDSNAAGLTKTSSNSLTRRFMHATVYLNTYYLTDPSYGYSQE RIVNTAE HELGHAIGLDH TNAVSV M QPAGSFYTIQPDDVQAVQKLYANNK Figure C.1. The protein sequence of L. reuteri ATCC PTA 6475 NT01LR0595 (Lr1291 homolog). The putative cleavage site is marked with two forward slashes (II); this site was predicted by SignalP 3.0 (3, 8). The putative active site is underlined, with the conserved residues in bold. The conserved methionine residue (“met-turn”) is also in bold. These similarities and sequence comparisons suggest that Lr1291 is part of a novel series of bacterial zinc-dependent metalloproteases of unknown function. Although there are other known bacterial zinc-dependent metalloproteases, none 179 of these proteins of known function were found to have significant sequence similarity. EXPERIMENTS AND RESULTS This section will briefly outline some of the experimental attempts to determine the function of Lr1291. Expression vectors and protein forms. Various expression vectors were used in attempts to over-express Lr1291. Original cloning attempts were made into traditional pET vectors, including pET-21b (C-terminal his-tag expression vector) and pET-19b (N-terminal his-tag expression vector). Due to the difficulty in cloning (owing to low transformation efficiencies and high background levels, a pET-21 b construct was never constructed; one pET-19b construct was constructed after multiple attempts), the Gateway system (Invitrogen) was employed. Several constructs were made successfully using this system, albeit at much lower efficiencies than suggested by the Invitrogen product manual. The vectors successfully used for constructs included: pET-DEST42 (C-terminal his- tag expression vector), pDEST-17 (N-terminal his-tag expression vector), and pDEST-15 (N-terminal GST-tag expression vector). Various forms of the gene were also used: the complete Lr1291 sequence, the Lr1291 sequence beginning with the putative cleavage site, and the complete Lr1291 sequence with the 180 active site mutated from HExxHxxGxxH to AAxxHxxGxxH. All constructs were confirmed by sequencing. Induction strains and conditions. Multiple strains were tested for induction with the various expression vectors. These included: E. coli BL21, E. coli Tuner, and E. coli Rosetta (DE3) pLysS cells (Novagen). Induction for these strains with the various vectors was attempted at 16° C, 25° C, 30° C, and 37° C in the presence and absence of 100 uM zinc acetate. Cell Iysates from all cultures were prepared by sonication and loaded onto SDS-PAGE gels. No difference was detected between proteins from uninduced and induced cultures under any conditions tested. Lysis of the induced cultures was observed on multiple instances, suggesting a toxic effect of the protein. Codon usage. Lr1291 contains multiple codons that are rarely used in E. coli (including four occurrences of AUA encoding isoleucine and six instances of GGA encoding glycine). In order to determine if codon usage played a role in the lack of high expression levels in E. coli BL21, Rosetta cells were employed for expression. E. coli Rosetta (DE3) pLysS cells from Novagen contain the pRARE plasmid, which supplies the tRNAs that recognize the following rare codons: AUA, AGG, AGA, CUA, CCC, and GGA (9). Use of Rosetta cells did not result in detectable expression levels of Lr1291. 181 .. 7.7 - . I ,0 Other systems for expression. When multiple attempts were made to express the protein in E. coli using various vectors, expression strains, and induction conditions, other systems were employed. These included an in vitro translation system (Qiagen EastPress Protein Synthesis Kit, catalog number 32501 ), over- expression in Bacillus subtilis (using the integrative pDR111 plasmid), and over- expression in Kluyveromyces lactis (New England Biolabs yeast expression kit, catalog number E10008). None of these systems resulted in detectable over- expression of Lr1291. When a construct containing Lr1291 was included in the in vitro translation reaction with the control vector, over-expression of the control protein (elongation factor EF-Ts) was completely shut down. This result suggests that production of Lr1291 may interfere with translation. Isolation from conditioned medium. After attempts at over-expression failed, experiments were carried out to try to isolate the protein from conditioned medium of L. reuten' ATCC 55730 and L. reuteri ATCC PTA 6475. For these experiments, both wild-type strains and mutants for each strain containing a disruption in Lr1291 were used. Cell-free conditioned medium from cells grown in deMan, Rogosa, Sharpe (MRS) broth (Difco) were used. Samples were collected from mid-log phase cultures, before and 15 minutes after exposure to bile, and from overnight cultures. The cell-free medium was concentrated under nitrogen, as some component of MRS broth clogged all types of centrifugation concentration devices tried. Despite using the same conditions where Lr1291 182 A“ 1.11 I, was shown to be over-expressed (15 minutes after bile exposure, Chapter 2) no consistent protease activity was identified in these samples. Because of the inconsistencies observed with concentrated conditioned medium from L. reuteri cultures, attempts were made to precipitate proteins from the cultures using 20% trichloroacetic acid. The samples were loaded onto SDS- PAGE gels and onto casein and gelatin zymograms. No difference in the secreted proteins was detected between the wild-type L. reuteri and the Lr1291 mutant strains. lmportantly, no protease activity was detected in any of the samples tested. The protocol for precipitation of proteins from L. reuteri conditioned medium is included at the end of this Appendix. Detection of activity. In all cases (protein expression and isolation from conditioned medium), samples were applied to either casein or gelatin zymograms to determine if any protease activity was detected. In several cases, protease activity was observed. Further investigation revealed that this protease activity was present in any E. coli lysate and was not specific to the production of the protein of interest. Note: Although some companies sell pre-made zymogram gels, the use of these products is not recommended. The results obtained with these products were highly variable. Due to the difficulty and inconsistencies in various zymogram 183 protocols, the protocol listed at the end of this Appendix obtained from Dr. Jenifer Fenton at Michigan State University is recommended for use. DISCUSSION The role of Lr1291, a protease of unknown function with homologs in various strains of host-associated lactic acid bacteria, should be investigated. The protein sequence shows intriguing similarity to eukaryotic matrix metalloproteases, a class of proteins known to play wide-ranging roles in normal development, inflammation, and the immune response. The MMPs have been implicated in important roles in the gastrointestinal tract, specifically in the intestinal immune response, the activation of defensins (antimicrobial peptides produced in the small intestine) and the development of inflammatory bowel disease (6, 7, 15). It is possible that proteases like Lr1291 may play an important role in the interaction of bacteria with the host. It is difficult to determine why there was a lack of experimental success for this project. The problems with the protein expression systems (low success rate in cloning, lysis of induced cultures, etc) suggest a level of protein toxicity, although the lack of success in over-expression of the mutant protein (active site changed from HExxHxxGxxH to AAxxHxxGxxH to inhibit activity) suggests that factors other than the protein activity are involved. A similar protein also found in L. reuten', Lr1017, did not cause lysis of the expression strains and was shown to 184 A' .'A‘ ’ Kind"? r. I be over-expressed using a pDEST-17 construct in the E. coli BL21 expression strain, suggesting that the problem is specific to Lr1291 and not a simple technical issue. The lack of activity observed with cell-free culture supernatant and precipitated secreted proteins from L. reuteri ATCC 55730 and ATCC PTA 6475 could be due to a low level of protein production under the growth conditions used or the use of an inappropriate substrate for this particular protease. It is also possible that the protein in question, Lr1291, is not actually secreted or may be secreted in an inactive form. Although casein and gelatin zymography are commonly used to detect general protease activity, it is possible that this particular protease has a more specific substrate. Several eukaryotic MMPs have either shown little or no activity against gelatin in vitro, thus the use of other substrates such as elastin, collagen, and various synthetic peptides should be considered for future use (4, 11, 12). Detection of activity from matrilysin (MMP-7) has been particularly difficult, and sensitive assays using synthetic fiuorogenic peptides as substrates have been employed (13). A screen of several of these synthetic peptides may be a more sensitive and possibly useful way of identifying protease activity from Lr1291. Other difficulties have been encountered in attempts to determine the role of various eukaryotic proteases. For example, MMP-23 did not demonstrate any 185 ,. " 1; n. ) protease activity against multiple substrates in vitro, suggesting either unique substrate specificity or a mis-folding of the protein during purification. Weak protease activity was finally achieved with a chimeric protein, combining the propeptide domain of MMP-19 (a metalloprotease known to autoactivate) with the catalytic domain of MMP-23, suggesting that activation of the protein (cleavage of the prodomain) may have been an issue in vitro (12). Despite the multiple attempts in this study at detecting activity from cultures of E. coli induced to express the protein and from conditioned medium of L. reuteri, mis-folding or a lack of proper activation of Lr1291 are possibilities that should also be investigated. Current work in the lab by another researcher suggests that some success in over-expressing the cleaved form of Lr1291 may be found using a Lactococcus lactis expression system. If this finding is confirmed, careful selection of a wide- range of substrates and sensitive assays should be employed for detection of protease activity. The value of the mutant strains of L. reuteri containing disrupted Lr1291 genes should also not be ignored. lf attempts to purify the protein are not successful, these strains could be employed in various assays, such as activation of defensins or production of cytokines and chemokines by eukaryotic cells, in attempts to determine the role of this protease. 186 PROTOCOLS OF INTEREST Protein precipitation from L. reuteri conditioned medium. 1. Grow L. reuten' in LDM-lll broth containing Tween 80 and glucose (MRS broth contains too many proteins; the background levels of precipitated proteins will interfere with results). At desired time, collect cells by centrifugation at room I} temperature for 10 minutes at 4000 rpm. Filter supernatant through a 0.22 (M filter into a 50 mL conical tube. I ’ 2. Add ice-cold 100% trichloroacetic acid (TCA) to a final concentration of 20%. Incubate on ice at 4° C (cold-room) for 48 hours. 3. Transfer mixture to Sorvall tubes and spin at 12,000 x g for 30 minutes at 4° C. Remove supernatant. 4. Resuspend pelled in 2 mL of ice-cold acetone (if starting with original culture volume of approximately 20 mL). Incubate on ice for 30 minutes. Transfer mixture to two 1.5 mL eppendorf tubes. 5. Spin at maximum speed in a microcentrifuge kept at 4° C for 30 minutes. Remove supernatant and air-dry pellets for 2 to 3 minutes. Resuspend pellet in desired sample buffer and proceed. Note: For zymograms, do not resuspend 187 pellets in the LDS sample buffer provided for use with the pre-cast Invitrogen protein gels. Although this buffer works well for visualization of the proteins, it will destroy any protease activity present in the samples. 188 u .u ‘Jfljl' a - I Zymograms (SDS-PAGE Substrate Gel Method) Stock Solutions - 30% Acrylamidelbis-Acrylamide (29:1) solution (purchase from Bio-rad, catalog #161-0156). Store in refrigerator. - 2.0 M Tris, pH 8.8. Store in refrigerator - 0.5 M Tris, pH 6.8. Store in refrigerator. - 0.625 M Tris, pH 6.8. Store in refrigerator. -10% 803 stock. Store at room temperature. -10% ammonium persulfate. Weigh 0.1 g ammonium persulfate in a microcentrifuge tube, add 1 mL of ddH20 the day of use, throw away when done. - Substrate - Gelatin: 2.0 mgImL Swine skin gelatin (Sigma, #G2500). Weigh 100 mg gelatin in a 50 mL conical tube, add 50 mL of ddH20, warm slightly to dissolve gelatin completely in water. Store in refrigerator. - Substrate - Casein: 2.0mglmL casein in 5mM Tris (Sigma, #07078). Dissolve 100mg of casein in 20 mL of 10mM NaOH and heat to get in solution while stirring. Neutralize by slowly dripping in 20mL of 10mM HCI while stirring on ice (sep funnel). Add 500 mM Tris, pH 7.5 (.5mL). Add 9.5 mL ddHZO. Store in refrigerator for no more than a month. *may not have to add the entire 20 mL of 10 mM HCI. Add slowly, stopping as soon as the solution becomes slightly cloudy (usually somewhere between 14 to 20 mL in my experience). Can make up any needed volume by 189 the addition of more ddH20. The addition of excess HCI will cause the casein to crash out of solution. - 4X Sample buffer (40% glycerol, 4% SDS, and 0.25 M Tris; pH 6.8). For 10 mL total volume, add in this order to a 15 mL conical tube: 0.4 9 SDS, 4 mL of 0.625 M Tris stock, 2 mL ddH20. Vortex. Add 4 mL glycerol and a small amount of powdered bromophenol blue. Vortex. - Running buffer (total volume of 4 mL). Add 1 L ddH20 and a stir bar to a 2 L graduated cylinder, weigh out 57.6 g glycine, 12.1 g Tris, and 4.0 9 SDS, add the three chemicals to the 2 L graduated cylinder, fill the graduated cylinder to the 2 L mark and stir until clear. Pour into a 4 L amber bottle. Add an addition 2 L of ddH20 to the amber bottle, cap, and shake. - Equilibration buffer - 2.5% Triton-X100 (total volume of 4 mL). Add 1.9 L ddH20 and a stir bar to a 2 L graduated cylinder place on a warm plate (medium setting) and stir, add 100 mL of Triton-X100 very slowly. Stir until clear. Pour into a 4 L amber bottle. Add an additional 2 L of ddH20 to the amber bottle, cap, and shake. - Incubation Buffer. 50 mM Tris HCI pH 7.5, 200 mM NaCl, 10 mM CaCI2, 10 uM ZnCI2, 0.02% Brij 35 (only good for one week once the calcium and zinc are added) - 4X Stain. Add 1 g Coomassie blue R250 to 1L methanol, stir and gravity filter. -1X Stain. Add to a 500 mL glass bottle, 100 mL 4X stain, 40 mL glacial acetic acid, and 260 mL ddH20, shake. 190 Procedure (Mini gels = 7.5 mL) Day 1 1. Substrate gel a. Mix the following in a conical vial for one gel: 2.0 mL 30% acrylamide stock 150 uL 10% SDS 1.4 mL 2.0 M Tris, pH 8.8 2.13 mL ddH20 1.86 mL gelatin stock or casein stock b. Add together simultaneously to mixture above: 83.6 ul ammonium persulfate 10.7 m TEMED 0. Mix pour immediately by slowly adding liquid gel to chamber with a transfer pipette. Slowly overlay gel with approximately 320 uL ddH20 (160 “L twice). d. Let stand at room temperature for 1 hour (remove water overlay before pouring stacker). 2. 4 % Stacking gel a. Mix the following in a conical vial for two stackers: 191 0.65 mL acrylamide 1.25 mL 0.5 M Tris, pH 6.8 50 mL 10% SDS 3.05 mL ddH20 D. Add together simultaneously to mixture above: 25 mL ammonium persulfate 6 mL TEMED c. Center comb in chamber and add liquid stacking gel with a transfer pipette, be careful not to create air bubbles. Let sit at room temperature for 30 minutes. 3. Loading the gel a. Enzyme samples are mixed with 4X sample buffer (32 mL sample: 8 mL sample buffer); load 40 (LL into well. b. For molecular weight ladders follow the protocol for each. c. Load samples with pipette tips while gels are completely submersed in running buffer, use a new tip for each sample. Check that the inner area is completely filled with running buffer and that the outer area is at least covered by buffer to the top black knob with running buffer. You can use a 192 transfer pipette to fill the inner area of the apparatus. This allows good current flow. 4. Running the gel Be sure that the red electrode is connected to the red outlet and the black to the black. Run the gel at 65 V until the samples are thru the stacking gel (approximately 30 minutes); then at 120 V (or 100 V if separation looks odd) until the blue line is below the gray seal at the bottom of the glass plates. Be sure that the protein does not run off the plates. While running the gel watch that the samples run at the same speed; watch for “smile” or “frown” sample lines. If they start to form, either increase or decrease the voltage to eliminate the “smile” or “frown”. 5. Preparing the gel for staining a. Once the samples are done, remove the gel apparatus from the buffer chamber set-up. Remove the gel from between the glass plates. Carefully remove the plates from their position on the gel apparatus. Gently lift one spacer (to release the vacuum that forms while running the gel between the plates) to remove the top plate (shorter plate). The gel will remain attached to the top (shorter) plate. b. Remove the stacker from the gel by pressing a piece of paper towel along the stacker portion of the stacker-gel junction. Next, gently pull up 193 on the paper towel to remove the stacker from the gel. Cut the upper right hand corner so that the orientation of the gel is known. By cutting the right top corner you are cutting the area where the marker is loaded or lane lot the gel. If more than one gel is run, out two (same side) or three corners so that the gels can be distinguished from each other. 0. Place each gel in a container, submerse each gel in equilibration buffer and cover container. Shake for 30 minutes in rocking water bath. Pour off old buffer in sink (be sure to retain gels in the container). Add new equilibration buffer to submerse each gel and cover container. Shake for 30 minutes in rocking water bath. Pour off buffer in sink (be sure to retain gels in the container). Add incubation buffer to submerse gel, cover container and incubate at 37C overnight (or for up to 48 hours). Day 2 6. Staining the gel Stain: gel for 1 hour with 1X stain: (completely submerse gel) at room temperature. Rock on rocker in covered container. 7. Destain Destain the gel in ddH20. 194 8. Storage of the gel Store gel in 10% glycerol or ddH20. 195 10. 11. REFERENCES Altschul, S. F., T. L. Madden, A. A. Schaffer, J. Zhang, Z. Zhang, W. Miller, and D. J. Lipman. 1997. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25:3389-3402. Bath, K., S. Roos, T. Wall, and H. Jonsson. 2005. The cell surface of Lactobacillus reuteri ATCC 55730 highlighted by identification of 126 extracellular proteins from the genome sequence. FEMS Microbiol. Lett. 5 253:75-82. = Bendtsen, J. D., H. Nielsen, G. von Heijne, and S. Brunak. 2004. Improved prediction of signal peptides: SignaIP 3.0. J. Mol. Biol. 340:783- 795. Lemaitre, V., A. Jungbluth, and Y. Eeckhout. 1997. The recombinant catalytic domain of mouse collagenase-3 depolymerizes type I collagen by cleaving its aminotelopeptides. Biochem. Biophys. Res. Commun. 230:202-205. Marchler-Bauer, A., and S. H. Bryant. 2004. CD-Search: protein domain annotations on the fly. Nucleic Acids Res. 32:W327-31. Medina, C., and M. W. Radomski. 2006. Role of matrix metalloproteinases in intestinal inflammation. J. Pharmacol. Exp. Ther. 318:933-938. Naito, Y., and T. Yoshikawa. 2005. Role of matrix metalloproteinases in inflammatory bowel disease. Mol. Aspects Med. 26:379-90. Nielsen, H., and A. Krogh. 1998. Prediction of signal peptides and signal anchors by a hidden Markov model. Proc. Int. Conf. lntell. Syst. Mol. Biol. 6:122-130. INovy, R., D. Drott, K. Yaeger, and R. Mierendorf. 2001. inNovations 12:1-3. Rawlings, N. D., F. R. Morton, C. Y. Kok, J. Kong, and A. J. Barrett. 2008. MEROPS: the peptidase database. Nucleic Acids Res. 36:D320- D325. Shapiro, S. D., G. L. Griffin, D. J. Gilbert, N. A. Jenkins, N. G. Copeland, H. G. Welgus, R. M. Senior, and T. J. Ley. 1992. Molecular 196 12. 13. 14. 15. cloning, chromosomal localization, and bacterial expression of a murine macrophage metalloelastase. J. Biol. Chem. 267:4664-4671. Velasco, G., A. Pendés, A. Fueyo, V. Knéiuper, G. Murphy, and C. Lopez-Otin. 1999. Cloning and characterization of human MMP-23, a new matrix metalloproteinase predominantly expressed in reproductive tissues and lacking conserved domains in other family members. J. Biol. Chem. 274:4570-4576. Welch, A. R., C. M. Holman, M. F. Browner, M. R. Gehring, C. C. Kan, and H. E. Van Wart. 1995. Purification of human matrilysin produced in Escherichia coli and characterization using a new optimized fluorogenic peptide substrate. Arch. Biochem. Biophys. 324:59-64. Whitehead, K., J. Versalovic, S. Roos, and R. A. Britton. 2008. Genomic and genetic characterization of the bile stress response of probiotic Lactobacillus reuteri ATCC 55730. Appl. Environ. Microbiol. 74:1812-1819. Wilson, C. L., A. J. Ouellette, D. P. Satchell, T. Ayabe, Y. S. Lopez- Boado, J. L. Stratman, S. J. Hultgren, L. M. Matrisian, and W. C. Parks. 1999. Regulation of intestinal d-defensin activation by the metalloproteinase matrilysin in innate host defense. Science 286:113-117. 197