ROLES OF THE MICROBIOTA IN INFLAMMATORY BOWEL DISEASES By Nabeetha Aarti Nagalingam A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Microbiology and Molecular Genetics 2010 Abstract ROLES OF THE MICROBIOTA IN INFLAMMATORY BOWEL DISEASES By Nabeetha Aarti Nagalingam Studying the roles of the microbial community structure as it relates to disease can give important insight into development of inflammation. This study employs concepts present in ecology to understand how altering bacterial communities in mammalian intestines affect inflammation at different stages of disease. The overall goal is to understand the role of the resident microbiota in initiation and progression of inflammatory bowel disease (IBD). Changes in the community can possibly act as a biomarker to diseases allowing preventative action to be taken or development of novel therapies. Work was done on two different models of IBD, dextran sodium sulfate (DSS)induced murine colitis and Helicobacter hepaticus triggered inflammation in IL-10/- mice (model of Crohn’s disease). Through culture independent techniques, changes in the microbiota were investigated, together with alterations in host mediators. In the first model, whether DSS-induced colitis was associated with changes in the microbiota was investigated. DSS administration was associated with microbial changes in intestinal communities in mice and these changes were ii present at onset of inflammation and persisted through later stages of disease. In the second model, whether changes in microbial community structure affected disease severity was investigated. The H. hepaticus triggered disease model was used to explore this question, and it was found that H. hepaticus infection of mice with different microbial community structures resulted in similar disease outcomes. This indicated that disease severity was independent from antibioticinduced changes in the microbial community in this model. We also show that the microbiota is essential in disease induction in mice in our colonies, as H. hepaticus monoassociated mice do not develop disease. These experiments demonstrate that the microbiota has important roles at several stages of disease, and these roles can be affected by other factors that influence IBD. In investigating stages at which the microbiota plays a role in IBD, we can better understand stages along disease progression when prognosis or treatment can be most effective. iii Acknowledgements I would first and foremost like to acknowledge my advisor, Dr. Vincent B. Young for giving me the opportunity to work in his lab. He has been a great mentor and I would like to extend my deepest thanks to him for guiding me through the years. I would also like to acknowledge my co-advisor, Dr. Thomas M. Schmidt, who took on the role of co-mentor when I re-located to the University of Michigan to continue my research. His support has made working across two campuses possible. The guidance of my advisory committee, Dr. Todd Ciche, Dr. Paul Coussens, Dr. Linda Mansfield and Dr. Martha Mulks, as well as collaborators Drs. John Y. Kao, Patrick D. Schloss and Gary Huffnagle, were invaluable. I would also like to thanks members of the lab, both past and present, for their support and friendship. iv Table of Contents LIST OF TABLES................................................................................................vii LIST OF FIGURES .............................................................................................viii Chapter 1 ..............................................................................................................1 The mammalian intestinal microbiota ...............................................................1 Introduction ........................................................................................................1 Effects of the microbiota on the host..................................................................2 Analysis of microbial communities .....................................................................7 Resisting and rebounding from change: Stability of the microbiota .................14 Influences on the microbiota ............................................................................17 The microbiota and disease .............................................................................25 Conclusion .......................................................................................................32 REFERENCES ....................................................................................................33 Chapter 2 ............................................................................................................46 Models of IBD.....................................................................................................46 Value of animal models....................................................................................47 DSS-induced colitis ..........................................................................................49 Helicobacter hepaticus infected IL-10-/- murine model of IBD.........................56 Conclusions......................................................................................................61 REFERENCES ....................................................................................................63 Chapter 3 ............................................................................................................72 Microbial ecology of the murine gut associated with the development of Dextran Sodium Sulfate-Induced colitis..........................................................72 Abstract and Key Words: .................................................................................72 Introduction: .....................................................................................................74 Materials and Methods.....................................................................................75 Results .............................................................................................................82 Discussion:.......................................................................................................93 SUPPLEMENTAL INFORMATION .....................................................................99 REFERENCES ..................................................................................................110 Chapter 4 ..........................................................................................................120 The effects of intestinal microbial community structure on disease manifestation in IL-10-/- mice infected with Helicobacter hepaticus. .........120 Abstract and Key Words: ...............................................................................120 Introduction: ...................................................................................................122 Materials and methods:..................................................................................124 Results: ..........................................................................................................131 Discussion:.....................................................................................................147 v Acknowledgements ........................................................................................152 SUPPLEMENTAL INFORMATION ...................................................................153 REFERENCES ..................................................................................................152 Chapter 5 ..........................................................................................................164 Summary, synthesis and future directions ...................................................164 Future Directions............................................................................................166 The role of the microbiota in initiation of IBD .................................................168 Conclusion .....................................................................................................174 REFERENCES ..................................................................................................175 APPENDICES ...................................................................................................179 Appendix A.......................................................................................................180 The effects of cefoperazone on Helicobacter hepaticus colonization. ......180 Purpose..........................................................................................................180 Methods and materials...................................................................................180 Results and conclusions ................................................................................182 REFERENCES ..................................................................................................190 Appendix B.......................................................................................................191 The effects of cefoperazone administration in H. hepaticus triggered disease in IL-10 defiecient mice. ....................................................................191 Purpose..........................................................................................................191 Methods and materials...................................................................................191 REFERENCES ..................................................................................................197 vi LIST OF TABLES Table 3.1 Showing distribution of OTU’s across treatments from major groups within the Firmicutes.....................................................................................92 Table 3.2. Changes in host gene expression following DSS treatment...............99 Table 3.3. Taxonomic distribution of rRNA genes clones from RDP classifier analysis of treatment group ........................................................................102 Table 3.4. Comparison of common terminal restriction fragment lengths generated by in silico digests of clones and the terminal restriction fragment lengths from T-RFLP via Msp1 digests. .....................................................109 Table 4.3. Fold change in expressions of host mediators from cecum tissue. All values are normalized to GAPDH expression and compared to cefoperazone untreated, H. hepaticus uninfected control group. Asterisk (*) denotes statistically significant changes (p<0.05). ...................................................154 Table 5.1 Roles of the microbiota in at various stages of inflammatory bowel disease. ......................................................................................................167   vii LIST OF FIGURES Figure 3.1. Histopathology in DSS-treated mice. Hemotoxylin and eosin (H&E) stained sections were prepared from cecum sections of (a) untreated control mice (b) mice after 3 days of DSS treatment and (c) after 14 days of DSS. Arrow indicates submucosl edema. H&E sections were also prepared from colon samples of (d) untreated controls (e) mice after 3 days of DSS treatment (arrow points to inflammation) and (f) animals after 14 days of DSS (arrow shows abscess). Histopathologic scores were calculated for sections from all 31 animals for (g) cecum and (h) colon sections. Statistical analysis done was Kruskal-Wallis test. *p< 0.05 **p<0.001. Initial magnification 40X. ........................................................................................83 Figure 3.2. Changes in host gene expression after 3-days (dark bars) or 14-days (grey bars) ± standard deviation of DSS administration. Expression levels were compared to expression in tissue from animals not exposed to DSS, after normalization with GAPDH. Statistical analysis done was Student’s ttest. Statistically significant differences (p<0.05) are denoted by asterisks (*). ......................................................................................................................85 Figure 3.3. Comparison of cecal communities in control animals (black triangles) and in animals following 3 days of DSS treatment (white squares) and 14 days of treatment (black squares). Using an OTU definition of 97% similarity, the Bray-Curtis similarity metric was calculated for each pair-wise comparison and then results displayed in denrogram format. Analysis by parsimony tests indicate that there is a statistically significant (*p<0.001) difference between the communities in control animals and in both groups of DSS-treated animals. ...................................................................................88 Figure 3.4 (A). Changes in the cecal gut microbial community following DSS administration at the phylum level. Analysis was done by ANOVA and statistical significance (p<0.05) is denoted by asterisks (*). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. ...........90 (B) Changes in the cecal gut microbial community following DSS administration within the phylum Firmicutes. Analysis was done by ANOVA and statistical significance (p<0.05) is denoted by asterisks (*). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. ...........91 viii Figure 3.5. Comparison of the cecal community in control animals (black triangles), and animals following 3 days of DSS treatment (white squares) and 14 days of treatment (black squares). Using T-RFLP, the Bray-Curtis similarity metric was calculated for each pair-wise comparison and then the results displayed in dendrogram format. Communities that were further analyzed, by construction of clone libraries, are indicated by arrows. Analysis by the parsimony test indicates that there is a statistically significant difference (*p<0.05 and **p<0.001). ...........................................................104 Figure 3.6. The rarefaction curves at 97% sequence similarity comparing the microbial communities in mice after 3-days of DSS treatment (pink), 14-days of DSS treatment (blue) and the controls (yellow). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. ............................................105 Figure 3.7. Comparisons between T-RFLP traces and histograms of the terminal restriction fragments (TRFs) of in silico digest of the clone library constructed from the control communities. ....................................................................106 Figure 3.8. Comparisons between T-RFLP traces and histograms of the terminal restriction fragments (TRFs) of in silico digest of the clone library constructed from the 3-day DSS administered communities. ........................................107 Figure 3.9. Comparisons between T-RFLP traces and histograms of the terminal restriction fragments (TRFs) of in silico digest of the clone library constructed from the 14-day DSS administered communities. ......................................108 Figure 4.1 Schematic of experimental designs showing (A) cefoperazone (Cef) and (B) vancomycin administration to IL10-/- C57BL/6 mice prior to or during infection with H. hepaticus (Hh)..................................................................126 ix Figure 4.2 Helicobacter hepaticus triggered inflammation in conventionally raised IL10-/- C57BL/6 mice compared to germfree counterparts. Representative sections of mouse cecum from experimental groups are depicted. (A) H. hepaticus uninfected conventionally raised mice appear normal. (B) H. hepaticus infected conventional (Conv) section shows edema, hyperplasia and inflammatory infiltrate. (C) H. hepaticus uninfected germfree control, and (D) H. hepaticus infected germfree cecum sections are both normal without signs of inflammation. (E) H. hepaticus forms close association to the intestinal epithelium, and in the crypts (yellow) compared to other microbes (red) in the lumen. (F) In germfree mice monoassociated with H. hepaticus, the bacteria are also found in close association with the epithelial layer (G) Quantification of indicators of pathology showing significant inflammation only in H. hepaticus infected conventionally raised mouse sections. Asterisk (*) denotes statistically significant changes (p<0.05). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. .........................134 Figure 4.3 H. hepaticus infection, as well as vancomycin treatment, significantly alters intestinal community structure. Asterisk (*) denotes statistically significantly different community structures calculated by parsimony test (p<0.05). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. ..............................................................................................136 Figure 4.4 Blinded scores showing significant inflammation in all H. hepaticus infected sections regardless of vancomycin treatment compared to uninfected controls. Asterisk (*) denotes statistically significant changes (p<0.05). .....................................................................................................140 Figure 4.5 H. hepaticus infection significantly alters the community structure of mice even after four weeks of termination of cefoperazone treatment. Asterisk (*) denotes statistically significantly different community structures calculated by parsimony test (p<0.05). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation....................................................142 Figure 4. 6 Blinded scores showing significant inflammation in all H. hepaticus infected sections regardless of cefoperazone treatment compared to uninfected controls. Asterisk (*) denotes statistically significant changes (p<0.05). .....................................................................................................144 x Figure 4.7 Changes in expression of host mediators in cecum tissue of mice administered cefoperazone and infected with H. hepaticus (p<0.05).........146 Figure A1.2 Cefoperazone concentrations above 0.25 mg/ml cause significant decreases in total bacterial loads and eradicates H. hepaticus. ................184 Figure A2.1 Experimental design showing H. hepaticus infection prior to cefoperazone treatment. ............................................................................192 Figure A2.2 Photographs of ceca removed from mice from four treatment groups. (Top left) Cecum is normal with luminal content of paste-like consistency, unlike cecum from mice infected with H. hepaticus that have thick tissue (top right). With cefoperazone administration (bottom panels), the cecum become distended and tissue fragile, filled with dark luminal fluid. For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. .........................194 Figure A2.3 show significant inflammation in mice infected with H. hepaticus. After administration of cefoperazone, inflammation was attenuated..........196   xi Chapter 1 The mammalian intestinal microbiota Introduction The human intestinal microbiota is comprised of seven of 70 known bacterial phyla: Firmicutes, Bacteriodetes, Actinobacteria, Proteobacteria, Verrucomicrobia, Fusobacteria and Cyanobacteria; and one of 13 archeal divisions: Euryarchaeota (1, 2). Of these, Bacteriodetes and Firmicutes are most dominant. This may suggest that communities are not very diverse (low richness and evenness), especially when compared to those in soils or oceans that have over 12 major phyla represented (3, 4), but this is far from the truth when genus or specieslevels are investigated. There are more than 400 species identified in the mammalian intestines, which were found to contribute to host development (5, 6). The participation of this microbial community is integral to our well-being and we are sometimes referred to as a “super-organism,” as both the microbes and host work in unison. This mutualism, however, is only maintained when the microbial community possesses a particular structure, composition, richness and evenness i.e. components of diversity. If this balance is perturbed, health can be compromised (7-9). Fortunately, studies on the diversity of the intestinal community demonstrate its resistance and resilience to external pressures (7, 10). 1 This review attempts to summarize roles of the mammalian gut microbiota in health and disease. As previously mentioned, a stable microbial community maintains health; therefore, how various factors change this structure will be discussed, as well as techniques used to measure these changes. Also, inflammatory bowel disease (IBD), a gastrointestinal condition, will be used as an example of a disease linked to changes in resident microbial communities. But first, I will address direct contributions of the microbiota to hosts in maintaining health and promoting proper host development. Effects of the microbiota on the host The microbiota is very active in molding its host. Gut microbes function to activate host genes in order to facilitate their own survival, as in the case of Bacteroides thetaiotaomicron activating production of defensins to kill potential pathogens and prevent competition for available space (11); or to benefit hosts via metabolism of previously unavailable nutrients such as butyrate (12). Microbial communities can also contribute to the host’s physiological and immunological development. Microbiota on host gut physiology There is a wealth of information on the benefits of intestinal microbiota on host development. A review by Wostmann summarizes a multitude of studies done on mapping differences between germfree animals and conventionally raised ones 2 (13). Some of these differences include increased ceca size, decreased intestinal weight and decreased colon surface area in germfree mice compared to their conventionally raised counterparts (14, 15). This might be due to the absence of butyrate-producing bacteria since this short chain fatty acid is the sole energy source for colonocytes, and lack of butyrate would lead to poor cellular development in germfree animals. Germfree mice also had less fat deposition seen post-mortem, indicating bacteria were important in lipid metabolism (16). Microbes contribute greatly to development and maintenance of healthy gut tissue. Intestinal architecture of germfree animals shows poor development of colonocyctes, as noted earlier. This is because these cells need the short-chain fatty acid, butyrate, as an energy source to proliferate, which is produced by the microbiota (17). A study also reported shorter crypt lengths and increased number of goblet cells that secreted mucus associated with the presence of a microbial community (18). Mucin is also very important in the barrier function of intestinal epithelium. Presence of a resident microbiota can stimulate production of polysaccharides and enhance the protective ability of the gut by providing a thicker layer between potentially harmful bacteria, or toxic components, and the host itself. Germfree mice also possess leaky tight junctions when compared to mice colonized with bacteria. Tight junctions are composed of a protein mesh that seals intercellular spaces in the epithelial layer. One study in particular shows that colonization with the probiotic Escherichia coli Nissle 1917 caused increased expression of the protein zonula occludens 1, involved in the formation of tight junctions (19). This leads to improved barrier function and protection against 3 invasion by pathogens or antigens, and underscores the contribution of “beneficial bacteria” to the host. Microbiota and the immune system The intestinal tract is an interface between hosts and their environments. As such, it is a location where cues are presented to the immune system from the external environment. Thus it is necessary that host’s defenses respond to appropriate signals; otherwise we would be constantly inflamed. It is seen, through work with germfree animals, that the intestinal microbiota is key in educating the immune system. Germfree mice were found to have fewer Peyer’s patches, thinner lamina propria, fewer plasma cells in germinal centers and fewer lymphoid follicles as reviewed by Round et al. (20). These mice also contained fewer T cells, paneth cells with reduced gene expression and B cells that produced less IgA compared to conventionally raised mice. All of these show importance of the resident microbiota in immune system development. Paneth cells are located in crypts and secrete defensins, small antimicrobial peptides. These cells also express RegIIIγ, which is involved in killing gram-positive bacteria. Use of antibiotics was shown to lower expression of this gene (21). This indicates that expression of this C-lectin may depend on the gut microbiota and emphasizes the degree of cross-talk between host and microbes in maintaining homoeostasis. 4 There are several mechanisms known for immunological responses elicited by pathogenic bacteria, such as effector systems on pathogenic islands like the Type III secretion system possessed by many pathogens such as Yersinia pestis, or flagella of Salmonella (22, 23). However, limited information is present for why resident bacteria do not trigger the host’s immune system and thereby can coexist with the host. Studies have cited various mechanisms that particular bacteria have evolved to suppress immunological responses. Many bacteria act on inhibiting NF-κB activation. Bacteroides thetaiotaomicron, for instance, is known to act down stream of TLR receptor signaling and NF-κB activation (24). Intestinal epithelial cells were incubated with Salmonella alone or Salmonella and resident microbe, B. thetaiotaomicron. It was found that with Salmonella alone, there was much less Rel A, the transcriptionally active subunit of NF-κB, in nuclei of host cells. This indicates that B. thetaiotaomicron is involved in export of this subunit and thus limits activity of NF-κB. Other studies show different mechanisms for tolerance of the indigenous bacteria (25, 26). Some of these are described by Cario et al. who reviewed tolerance of intestinal immunity through toll-like receptors (TLRs) (27). Consequently, the host is not simply blind to the presence of indigenous bacteria, but employs a two-way communication that results in tolerance. Microbiota on other locations besides the gut As discussed above, gut microbiota is key in proper development of adaptive and innate responses against pathogens and irritants. Therefore, it is not surprising 5 that the microbiota would also have far reaching effects in host organisms, and not just on the intestines in which they are located. It is known that signals from the brain have effects on the microbiota, but the converse is also true, the microbiota has an effect on the brain’s function as well. Vagus nerves, for instance, can trigger vomiting during food poisoning (28). Studies with germfree mice showed that these animals had an increase in levels of adrenocorticotrophic hormone (ACTH) and corticosterone, during a particular stress experiments, compared to specific pathogen free controls. This study indicates that the gut microbiota may have a role in suppression of ACTH and corticosterone production (29). A study was also conducted investigating whether there was a connection between the microbiota and pain. The authors reasoned that the perception of pain was necessary for organisms to adapt to their environment and external stresses. They conducted experiments using germfree mice and found that a microbiota was indeed necessary for proper development of the response to inflammatory pain (30). The gut microbiota was also shown to play a role in liver functioning, as well as with deconjugation of bile acids (by some Lactobacilli), and insulin resistance (31). Dumas et al. conducted a study on BALB/c mice to study the metabolic profile associated with insulin resistance and found that metabolites indicative of this condition was related to host diet (31). They saw that the metabolic phenotype associated with disease could be correlated to amounts of fat in the diet. By extension, since the microbiota plays a large role in lipid available to the host, the 6 gut’s microbial community plays a role in altering host metabalomics and occurrence of certain disease (32). To underscore the far reaching effects of the gut microbiota, one recent paper also reported effects of this community on lens of the eyes (33). Lipid content of lens and retina of eyes, from mice with a gut microbiota, was measured together with those from germfree animals. They found that conventionally raised mice had less phosphatidylcholines in their lens indicating exposure to oxidative stress. It is thus clear that the intestinal microbial community affects more than digestion. Analysis of microbial communities Earlier, the importance of community diversity and stability on the maintenance of health was briefly mentioned. Before discussing the nature and impact of specific changes in the microbial community, I will first summarize techniques used for measuring these changes. Tools and techniques For a long time microbiologists studied organisms by growing them in culture. However, more than 80% of intestinal bacteria cannot be cultivated using current techniques, as described by “the great plate count anomaly” which addresses differences between the greater number of microbes seen with a microscope than those actually grown on culture plates (34). When Carl Woese introduced the 16S rRNA-encoding gene as a target to study microbial phylogeny, new doors opened for investigating microbial diversity (35). 7 Ashelford et al. have described features of the 16S rRNA gene that makes it appropriate and useful for phylogenetic classification (36). This gene is highly conserved given its function in protein synthesis; however, nine variable regions interrupt this conserved gene. The first characteristic allows us to design broadrange primers that bind conserved regions so that amplicons can be retrieved that include many groups of microbes. The second feature allows these amplicons to be distinguished from each other since aligned sequence of these variable regions will differ from gene to gene, and thus bacterium to bacterium. Many molecular techniques used for studying diversity within the microbial community depend on these features of the 16S rRNA-encoding gene. Some of these will be described below. Methods used to study the microbiota include denaturing gradient gel electrophoresis (DGGE)/ temperature gradient gel electrophoresis (TGGE), terminal restriction fragment length polymorphism (T-RFLP), fluorescent in-situ hybridization (FISH), clone libraries, and more recently, high throughput pyrosequencing. In DGGE, the 16S rRNA genes from a sample are amplified through PCR and 16S rRNA gene amplicons are run across a polyacrylamide gel that has a vertical gradient of denaturing substance. As the double stranded amplicons travel down the gel, they pass through changing concentrations of this denaturing substance and the double stranded helices dissociate at various points. Since each helix is composed of DNA that spans a hypervariable region, these strands would have different base pairs and hence separate at different points along the gradient. The 8 resulting gel would have a ladder-like appearance. If amplicons from two different communities, of differing community structure, were run side-by-side, it would be expected that patterns of “rungs” of the ladders would be different. Further analyses are normally done to identify microbes according to patterns on the gel. For example, hybridization analysis with species-specific probes can be used to identify specific microbes (37). Bands from the gel can also be excised and sequenced in order to ascertain microbial identities. An alternative approach involves using a temperature gradient (TGGE) instead of a denaturing gradient, to allow double stranded DNA helices to denature along the gel (38). T-RFLP is another fingerprinting technique. Here PCR-amplified 16S rRNA genes of a bacterial community are digested with restriction enzymes and cleaned product is visualized using spectroscopy. This visualization is possible because terminal fragments are fluorescent as one of the primers, either forward or reverse, has an attached fluorophore (e.g. 8-carboxy-fluorescein label) (39). Using T-RFLP, relative abundance of different operational taxonomic units (OTUs) can be seen, as intensity of light emissions is proportional to abundance of each OTU, and different OTUs are designated by peaks generated at cut sites (39). This makes the use of traditional macro-ecological indices (e.g. Shannon or Simpson Indices), employed to measure richness and evenness, easy to apply. As mentioned briefly above, sequence-specific probes can be used to investigate microbial identities using hybridization techniques. FISH is commonly used for a quantitative measure of particular bacteria in an environmental community, such as the gut, in situ (40, 41). In this technique, a fluorescently labeled 9 oligonucleotide is used to bind to genes of interest (frequently the 16S rRNA gene) of microbes in a community. This oligonucleotide sequence would have a certain degree of specificity according to its design, that is, compliment a variable region for species-level detection or a more conserved region for higher phylogenetic resolution such as phylum-level detection (40-43). The labeled microbes can then be visualized by microscopy or flow cytometry. Another method used to study the microbial communities is construction of clone libraries. Clone libraries are typically constructed from full-length 16S rRNA gene amplicons using primers that bind to conserved regions (10). The amplicons are then ligated into vectors and transformed into a host cell, typically Escherichia coli. These cells, carrying individual 16S rRNA gene fragments are cultured to retrieve sufficient copies of the gene that are then sequenced. These sequences are then aligned, and assigned identities by comparing to known bacterial sequences that are already archived. This technique allows researchers to study communities by comparing who is there and in what relative abundance. Although this is a very powerful technique for analyzing a community, it is limited in that there is relatively little sampling effort exerted (44, 45). Here, pyrosequencing has bridged the gap with deep sampling, with hundreds of thousands of sequence reads per run compared to only about a hundred sequences with clone libraries (46, 47). In pyrosequencing, a hypervariable section, about 300 base pairs, of the 16S rRNA gene is amplified from a community. Adaptors are ligated on either end of PCR fragments, which aid in community separation in future analyses, and 10 facilitate binding to beads. Beads are captured in droplets that house PCR reactions so that there are millions of DNA strands associated with each bead. Droplets are then broken and the beads placed into a microtitre plate, along with enzymes. Sequencing occurs by measuring emissions generated when deoxynucleotides wash over the plate (47, 48). This techniques generate massive amounts of data and allows investigators the opportunity to observed the “rare biosphere” (49). These methods described above have all been invaluable in studying gut communities. DGGE was first reported in a 1993 publication where it was used to investigate microbial communities from various locations from microbial mats and waste water samples (37). These authors used PCR to amplify the third variable (V3) region of the 16S rRNA gene and used this amplicon to conduct the DGGE. Zoetendal et al. also used this technique to study the microbiota of the gut by comparing community composition of feces and tissues from different locations along the colon (50). Likewise, through T-RFLP, a broad overview of the community was observed when Keuhl et al. investigated whether there were changes in community richness and evenness after infection with Helicobacter hepaticus (39). The authors noted that presence of H. hepaticus had minimal effects on other members of the community by comparing changes in resulting traces. Although these techniques present information on general differences between communities, they do not provide identities of members in the community. Supporting techniques are used for this purpose, such as sequencespecific probe hybridization in DGGE that would bind to DNA of certain members 11 of the microbiota, or in silico digests of known bacteria to generate predictive traces to compare with T-RFLP data. FISH has the advantage of showing identities of certain groups in a particular location. Franks et al. studied changes in abundance of certain bacteria in fecal microbiota of nine participants over eight months (40). This hybridization technique provided rapid results that were quantitative and allowed investigators to track certain species of interest, Bacteriodes fragilis and Bacteriodes distasonis among others. One constraint however, is since bacteria are measured using emissions from fluorophores attached to probes, there is a limit to the number of probes that can be used. This issue can be overcome using sequencing methods. These methods allow generation of data that have components of relative abundance, richness and identity of community members. With pyrosequencing, communities can be sampled to such an extent that previously rare members are observed. Dethlefsen et al. used this method to study changes in the human microbiota induced by antibiotic treatment (49). They identified more than 5000 taxa associated with the human microbiota, and were then able to further analyzed communities by focusing on specific groups, including the Bacteriodes, Clostridiales and Lachnospiraceae. At the moment, this method is more expensive than others previously mentioned, however, the volume of data generated makes this technology very appealing. 12 Parameters in measuring microbial diversity To begin to understand microbial communities, data need to be sorted and interpreted. There are two important parameters that are measured in studying diversity of intestinal microbial communities: richness and evenness. Richness involves enumerating different bacteria that are present and is represented by numbers of operational taxonomic units (OTUs) in communities; therefore, defining what makes each OTU different is necessary. In T-RFLP, an OTU is defined by each peak in the trace generated; or in clone libraries, the designation can be applied after 16S rRNA gene sequences are aligned and compared. Sequences that are 97% similar to each other are generally considered to be of the same species (6). This is still subjective since some authors prefer to use more stringent criteria for defining “species” such as 99% similarity (5). Once cutoff levels are defined though, analysis of communities is quite powerful and represents an accurate and easy way of measuring diversity. Evenness refers to relative abundance of groups within communities. This can be measured using quantitative methods such as clone libraries, pyrosequencing, real-time PCR, FISH, and microscopy. Fingerprinting methods, such as T-RFLP, also gives data on relative abundance by noting relative fluorescent units associated with each peak. This method is relatively cheap, and useful in retrieving a broad overview of changes in a community. It is becoming clear that relative abundances of certain groups, and not merely their presence or absence, is key in onset of disease (10, 51). Two previous parameters, richness and evenness, in part, describe structure of the community. Another feature of microbial communities is that of function, 13 and depends on community composition. Composition refers to identities of the various bacteria present and can be measured using sequencing methods. Denoting membership allows investigators to potentially assign function and thereby begin to understand mechanisms of community dynamics. Resisting and rebounding from change: Stability of the microbiota We have described how to measure community structure and composition. This structure is subject to shifts that may be due to a number of factors. Ecologists have been considering changes in terrestrial and aquatic communities for decades and have generated concepts to explain and predict changes in these communities. Microbiologists have now applied these concepts in studying the microbial world, be it in soil communities or gut microbiota (3, 10). Intestinal microbial communities experience many perturbations, including infection by pathogens or treatment with antibiotics. In studying communities’ responses to these disruptive forces, one must consider resistance and resilience, two key ecologic concepts developed for studying communities of macroorganisms. Resistance is the ability of a community to maintain its structure under stress, while resilience is its ability to return to its pre-stress state after brief change in structure (52). These two terms are associated with stability of a community, a condition thought to be necessary for health since alterations in diversity of gut microbial communities have been believed to play a role in some diseases such as inflammatory bowel disease (IBD) and antibiotic associated diarrhea (10, 53). 14 The question of whether diversity begets stability has been ongoing for at least the past 50 yrs (54). Ecologists first believed that more diverse communities are more stable. This was supported by work on terrestrial communities from Odum (55), Elton (56) and Tilman (57) who thought that simple communities were susceptible to perturbations that left these communities open to invasion. MacArthur had similar views after studying predator-prey interactions (58). More recently, however, this view was challenged. Robert May used mathematical models to show that diversity does not lead to stability in a community (59). In fact, he showed that increased diversity destabilized the community. This issue was further complicated when looking at food web relationships where strength of associations also played a major role in the stability of community structure. In these cases, it was seen that observations noted by Odum, Elton and MacArthur did not apply since there was more than one trophic level involved (55, 56, 58, 59). Microcosm experiments show that there is a positive correlation between diversity and stability. This is seen at community-level (different groups of bacteria) and not population-level (within a particular group, be it at phylum or genus-level). At the population-level, it was seen that increasing diversity did not affect community stability. Findings from these experiments led scientist to note that two explanations were possible in explaining community stability. First, increasing diversity increased the chances of variable species response to a perturbation and hence some species can help maintain the normal functioning of the ecosystem. Secondly, increasing diversity increased chances of functional 15 redundancy so if certain species are lost during a disturbance, communities still function as all the tasks are still occurring (60, 61). These two explanations give rise to the “insurance hypothesis.” It is also noteworthy that diversity is not the “driver” of stability, but merely shows a positive correlation with it. Mechanisms by which this occur are still under investigation. With this hypothesis, it is thought that in the event of a perturbation, a community either shows resistance, in that it generally does not show great change, or it is resilient, and bounces back after an initial change because hardier members of the community compensate for decrease of susceptible ones until they recover (61). The complexity of this issue is emphasized by contradictions observed as one study showed that a less diverse community was more resistant and resilient than highly diverse one (62). Authors explained that relationships between diversity and stability in communities depend on pre-stress states of those communities and mechanisms that drive them. In considering the gut microbial community, the same ideas apply. One would expect that diversity correlates with stability at the community level. As seen with some of the antibiotic work, communities can be resilient (10, 46). And as suggested by the insurance hypothesis, this resilience may be due to functional redundancy in communities. Functional redundancy is known to be present in our gut microbiota, for instance several members are able to produce butyrate (12, 63). 16 Influences on the microbiota There are many factors that sculpt microbial community structure in the intestine and, on occasion, cause disruption. From birth, when colonization occurs, the microbiota is shaped according to its anatomical location as conditions (pH, oxygen concentration) vary, diet changes, pathogens infect and antimicrobials are administered. It is not surprising then that there is great inter-individual variation. However, there are also great similarities due to evolutionary commonalities among vertebrates that give rise to the idea of a “core” microbiome (6, 64). Microbiota changes with age The fetal environment is thought to be sterile and colonization starts during birthing. Colonization, thus, depends on both the method of birthing (caesarian section versus natural child birth) (65), and whether or not the child is breast-fed (66, 67). This early microbiota then evolves into a mature community that begins to stabilize soon after weaning. This community is then influenced by diet, antibiotics and other environmental factors that contribute to inter-individual variation, and persists into old-age. Palmer et al. investigated development of the intestinal microbiota by following infants through their first year of life (68). They assessed colonization through stool sampling and comparisons with stool samples from other members of the family. Mothers’ milk and vaginal swabs were also investigated in an attempt to map route of colonization. The authors found that the gut microbiota in the first days to months of an infant’s life is variable and is related to environmental 17 exposure, i.e. vaginal route, breast-feeding. The microbiota of these 12 infants, however, converged toward the end of their first year. This community resembled that of an adult, consisting of an abundance of Firmicutes, Bacteriodetes, with some Verrucomicrobiae and Proteobacteria. Contrary to many publications (69, 70) though, there was a low prevalence of Bifidobacteria. The authors suggest that previous papers may have exaggerated the importance of this group due to its probiotic effects, and lost focus on other more prevalent groups (68). Once the community reaches this stabilized adult structure, fluctuations from lifestyle, e.g., diet and disease, are its main effectors. This was demonstrated in another recent investigation involving more than 35 000 adults (71). It was found that bacterial communities, in stool samples, were generally the same over age and gender. A few individual groups such as Enterococci, increased with age while Bacteriodes decreased. Bifodobacteria and Lactobacillus appeared to be stable. These data were retrieved by culture so do not represent species that may be difficult to grow via methods used. Other limitations included the lack of knowledge of antibiotics taken or diet of participants. Diet is known to play a role in the structure of gut microbiota and these authors suggest that change in bacterial communities with age could be due to changing diets and not the actual process of aging (72, 73). 18 Microbiota changes with location Studies often use fecal samples in analyses (68, 73). Although, stool is convenient and non-invasive, it may not be representative of the gut microbiota since microbial communities are not homogenous along the gastrointestinal tract, or across the lumen. Gut microbiota is divided into two groups, autochthonous (commonly referred to resident communities that are closely associated to the host) and then allochthonous (transient bacteria that do not get permanently associated with host and may include pathogens) (74). In studies investigating resident microbiota, the autochthonous community can be investigated using biopsy samples, which capture mucosal microbes that interact intimately with a host. It was also found that certain resident bacteria associate with hosts at particular locations along the intestine, at varying abundances (75). The alimentary canal, although continuous, harbors various environments from mouth to anus (51, 75). Focus will be placed on the intestinal section since much emphasis has been placed on this community for its role in host development and maintenance of health (1, 12, 76). Differences in anatomy between intestinal sections are obvious with the cecum being a blind end, while the colon is tubular. Other differences are detailed in a review by Snipes (77). There are also differences in the immune elements among different regions of the intestines (78), such as Paneth cells being present in small intestines and not colon. These characteristics may contribute to the variation seen along different sections of the intestinal tract. Wang et al. (75), through molecular techniques, saw that human jejunum had the lowest microbial richness compared to distal ileum, ascending 19 colon and rectum. By comparing sequences against those of known bacteria in the database of the National Center for Biotechnology Information (NCBI), they also found that the main difference was that the jejunum had the least Bacteriodetes but the most Streptococcus (68%) and Proteobacteria compared to other three regions. Another group suggests that acid-tolerance by Streptocococcus may select for this bacterium in the upper intestine since this environment has a low pH (79). They also showed a complete lack of detection of Clostridium XIVa group in the jejunum while other parts had between 20 and 33% prevalence. The ileum and rectum both had a high prevalence of Bacteriodetes (49 and 44%, respectively). Previous work by this group showed a gradient of increasing load and richness from terminal ileum to colon in a single individual (80). However, Zoetendal et al. (50), using DGGE, noted that bacteria along colon mucosa of 10 individuals were evenly distributed. Wang et al. (75) also showed similar distribution except in the case of Bacteriodetes. Since Wang et al. (75) studied a single subject while Zoetendal et al. (50) had 10 participants, this difference could be due to variation noted among individuals, as well as the sampling methods employed. Certain microbes may also be associated with different regions along the crosssection of the lumen since different microenvironments are present; for instance, anoxic conditions exist at the center of the lumen particularly within stool (81), whereas microaerophilic conditions are present near the vasularized epithelium (82). 20 Communities retrieved also depend on samples and sampling techniques since there are differences in dominant bacteria in feces and those attached to mucosa as observed through TGGE, for instance (8). However, fecal and biopsy communities look more similar to each other in one individual than two fecal samples from two different individuals due to inter human variability (5, 83). We can even go further in describing variation among mammals where it was seen that grouping not only depends on genetics but also diet (64). Microbiota changes with diet Diet has been known to influence gut bacteria since it selects for certain groups that can carry out certain functions, such as Bacteroides to break down protein or Clostridia to act on complex carbohydrates in fiber degradation. Therefore, one would expect a predominance of Clostridia in an individual consuming a vegetarian diet. This was supported in a study done by Hayashi et al. (72) where gut microbiota of a female vegetarian was investigated and compared to those of three healthy men. It was found that the vegetarian had a high proportion of Clostridium XIVa and Clostridium IV, as well as Clostridium ramosum from cluster XVIII. This latter bacterium may be important in individuals on a vegetarian diet since Finegold et al. also reported that this microbe was present in 52% of vegetarians sampled in their study (84). Actual function, however, is still under investigation. Researchers have also found that Bacteriodes were higher in individuals with a greater meat intake. In addition, Bacteriodes convert bile into products that are known carcinogens and thus a high proportion of this group has 21 been linked to colon cancer (85). This has been the subject of debate as a vegetarian diet is proposed to reduce chances of developing colon cancer, emphasizing a link between diet, microbiota and disease. Thus, diet is not only associated with shaping gut communities, but also selecting microbes that are associated with development of disease. Microbiota changes with a pathogen Pathogens are organisms that cause disease. Many, such as Clostridium difficile and Salmonella enterica, are linked to intestinal disease in humans, and are associated with shifts in resident microbial communities (7, 51). Studies on these organisms show that infection normally occurs after shifts in community structure, due to antibiotic treatment, and implies deterioration in colonization resistance. Salmonella enterica Serovar Typhymurium infection will induce a typhoid-like illness in mice, i.e. mice develop a disease resembling human typhoid fever, with temperatures as high as 104°F and non-bloody diarrhea. As a human pathogen, this microbe induces enteritis. FvB mice are susceptible to Salmonella infection without antibiotic pretreatment. This provided a model to observe alterations of gut microbiota in the presence of a pathogen. Upon infecting mice with Salmonella, it was found that the diversity of the resident microbial community changes (86). Salmonella colonized cecum and colon better than distal small intestines, as shown by qPCR data. In the colon, numbers actually decreased after 1 week compared to the 3 days post infection. This colonization coincided 22 with changes in load of other bacterial groups. Bacteriodetes decreased in the cecum and colon. Lactobacillus/Enterococcus decreased significantly in the distal small intestine after 3 days Salmonella infection; and the Eubacterium rectale/Clostridium coccoides group decreased in all sites measured, with a corresponding increase in Clostridium perfinges. However, these changes could be due to host responses to infection. Actual onset of diarrhea can only be associated with the 1 week post infection group since no diarrhea was seen 3 days post infection. Once Salmonella was cleared, the microbiota rebounded to it previous set point. The authors observed only certain groups of bacteria to be indicative of changes in the microbiota. It should be noted that there may be other changes seen in groups that are not represented by the primer sets used. A more wholistic approach, using “universal” primers, would be more informative in probing community changes more thoroughly (10, 46). However, the study shows subsequent shifts in the microbiota due to presence of a pathogen. Although this change in the microbiota may not be directly caused by Salmonella invasion, it is definitely associated with onset of diarrhea. Similarly, a study by Chang et al. showed that patients recovering from C. difficile infections had gut microbial communities more similar to healthy participants, than to those with active disease (7), indicating a return of a “healthy microbiota” in recovering patients. C. difficile infections are normally treated with an antibiotic regime, which does not only target this pathogen, but also members of the resident community. This introduces another agent of perturbation to gut microbial communities. 23 Microbiota changes with antibiotics It has been shown that antibiotics can alter intestinal murine microbial communities (51, 87). In so doing, colonization resistance is lowered and infection by a pathogen such as Salmonella is very efficient (51). Antibiotics have long been recognized to significantly altering the gut community (88). Van der Waaij et al. touted the use of antibiotics in “decontaminating” the digestive tract since they saw dramatic reduction in aerobic and anaerobic bacteria after treatment with various drugs (88). This survey was done by culture methods thus many bacteria would have been undetectable (34). Never the less, antibiotics can greatly impact gut communities. Antonopoulos et al. (46) showed that degree of perturbation depended on specific treatments used. Researchers, through pyrosequencing technology, showed that cefoperazone induced long-term reduction in community diversity (up to six weeks after cessation of treatment). Another drug combination, amoxicillin/metronidazole/bismuth (AMB), only showed short-term reduction in community diversity with richness being restored after only 2 weeks after antibiotic treatment stopped. This exemplifies elasticity of community diversity under certain conditions. Previous work from this group also showed remarkable resilience of some members of the gut microbiota after a perturbation caused by an antibiotic regime (10). Although the antibiotic administered was for treatment of sinusitis, the gut microbiota of the patient in this study showed shifts among different microbial groups. This change in diversity of the gut community was associated with diarrhea. On completion of treatment, diversity of the microbiota resembled that of the community before antibiotics. 24 Bowel movements also returned to normal. This suggests that certain microbial diversity is associated with health, and that disturbing it can lead to disease. Alternatively, health may be attributed to overall community structure with a predetermined balance perpetuating a healthy state. The microbiota and disease The microbiota contributes tremendously to health and development of its host, as reviewed above. We have also seen that this community is very resilient. But what happens when the microbiota is altered and cannot compensate for associated changes? In some cases, this community disturbance has been associated with onset of disease. Inflammatory bowel disease Inflammatory bowel disease (IBD) includes both Crohn’s disease (CD) and ulcerative colitis (UC). The former is characterized by lesions, which may occur anywhere along the gastrointestinal tract. These lesions can penetrate through layers of the GI tract, and may even corrode through the gut wall and create fissures into other organs such as the bladder. The latter is normally confined to the lower intestines and rectum, and lesions are more superficial, affecting the epithelial layer and lamina propria (89). Since no specific bacteria can be linked to the development of IBD in humans, Koch’s postulates cannot be fulfilled, leading to the current view that this disease 25 is a result of dysbiosis (changes in the microbial community structure). Koch’s postulates are as follows: 1. The microorganism must be found in abundance in all organisms suffering from the disease, but should not be found in healthy animals. 2. The microorganism must be isolated from a diseased organism and grown in pure culture. 3. The cultured microorganism should cause disease when introduced into a healthy organism. 4. The microorganism must be re-isolated from the inoculated, diseased experimental host and identified as being identical to the original specific causative agent. Koch’s postulates are especially challenging to fulfill in the context of intestinal disease since many microbes are difficult to culture by traditional methods (32), therefore may not be easily isolated. Even if they are isolated, interactions with other microbes or host may be required for growth, preventing pure culture. However, many studies have been cited that link the microbiota with IBD (90, 91). IL-10-/- mice have been convenient in showing roles of microbes in onset of IBD. Since this anti-inflammatory cytokine is absent in these mice, inflammation perpetuates without the inhibitory force of immune regulation that wild type animals possess. Disease is then mediated from a Th1 type response. It was found that wild type conventionally raised or germfree mice did not develop colitis, neither did germfree IL-10-/- mice (92). However, IL-10-/- conventionally raised mice developed disease leading authors to suggest that a resident intestinal 26 microbiota was necessary for development of colitis in these immune altered mice (92). Also, it was found that disease was only present after mice were gavaged with fecal slurry and not sterile filtrate of fecal slurry or sterile bacterial lysates, indicating that microbes are responsible for immune stimulation (93). Alternatively, it was postulated that one mechanism by which resident bacteria cause colitis is by compromising the colonic epithelial barrier (94). Ohkusa et al. showed that different indigenous bacteria were capable of inducing varying levels of cytokine production from the host intestinal tissue (94). Activity of these inflammatory molecules suggests that resident microbes could induce and maintain inflammation and perhaps, did not cause disease in vivo since host mediators kept them at bay. The host thus appears to control intestinal bacteria from invading, emphasizing the tug-of-war that occurs in this dynamic system. Nonetheless, resident microbes have an important role in onset of IBD. Animal models of IBD IBD is of unknown etiology. There are several animal models available that mimic appropriate immunological and histopathological responses. Although these models do not encompass all the complex parameters involved in human disease, they have been used to elucidate pathophysiology of disease, and have also been useful in determining possible therapies. There are two major types of animal models used to study IBD: chemically induced (dextran sodium sulfate, trinitro-benzene sulphonic acid) or immuno-deficient (T cell adoptive transfers, IL10-/-). In the following, the dextran sodium sulfate (DSS)- induced colitis model, 27 as well as the Helicobacter hepaticus infected IL-10-/- mouse model are discussed. These are frequently used models employed by our laboratory in studying IBD related changes in the microbiota (39, 95). Dextran sodium sulfate (DSS) is commonly used as a means to induce colitis reliably (96). This chemical is heparin-like in nature and too large to transit intestinal walls (97). This chemical can induce two types of disease according to administration, acute or chronic. Acute disease occurs after one time dosage at about 3.5% DSS, while chronic colitis occurs at a lower dosage that is repeated at intervals (98). Mechanisms by which it causes diseases is still under investigation (99). There has been vast work done on effects of DSS on host tissues and immune response as it relates to ulcerative colitis (UC) (100, 101). Using this model, Medzhitov and colleagues have shown that TLR 4 knockout mice did not develop colitic lesions on treatment with this chemical (102). MyD88 knockout mice also fail to develop characteristic lesions of UC (25). More recently, Ungaro et al. found that TLR 4 antagonist antibody ameliorates inflammation in colitic mice (103). These studies suggest that intestinal bacteria can play a role in development of UC via the MyD88 pathway. Another model of IBD linked with the microbiota is infection of IL-10-/- mice by Helicobacter hepaticus (104). There have been several studies showing development of colitis in IL-10-/- mice infected with this microbe (95, 105, 106). H. hepaticus induced colitis is a T cell response (104). Recent evidence has also 28 linked Th 17 that produces IL-23 and INFγ in development and modulation of this infection (107). The mechanism by which this bacterium causes colitis is unknown; however, it has been found that H. hepaticus has a pathogenicity island (PAI) that is involved in induction of disease (106). This PAI was involved in significantly increasing disease of the cecum, as seen through histology and proinflammatory mediators, of B6.129-IL10tm1Cgn (IL10-/-) mice compared to those infected with an isogenic mutant (106). The PAI region investigated showed immuno-regulatory effects by showing lower IgG2c and IgG1 responses than wild type, but did not affect colonization ability. Another paper from this group also showed that the B component of cytolethal distending toxin (CdtB) was important in intestinal colonization. Again, there were also lower levels of IgG2c and no IgG1 response indicating that CdtB was involved in immuno-regulation (105). Therefore, for this bacterium, PAI and CdtB are important in establishing inflammation and causing colitis. It is important to note that H. hepaticus induced colitis only occurs in mice containing a conventional microbiota. Dieleman et al. has shown that disease only ensues in the presence of a resident microbiota and not in H. hepaticus monoassociated IL-10 deficient mice, on a mixed (C57BL/6 x 129/Ola) or inbred (129/SvEv) genetic background (108). It should be noted that germfree animals are not “normal” in that they do not have a developed immune system and would not react as conventional animals. Also, disease does not occur in mice with an intact immune system since several studies with this bacterium in wild type animals have not resulted in inflammation (39). As previously discussed, a 29 pathogenicity island facilitates onset of disease, and CdtB is needed for establishment (95, 105). In this model, disease then seems to depend on several factors: resident community, host and H. hepaticus itself. Probiotics Non-resident bacteria can be used to enhance or rescue deficient functions of the gut microbiota so that disease is prevented or ameliorated (109, 110). Probiotics is defined as “living microorganisms which when administered in adequate amount confer a health benefit on the host (17).” These benefits can be conferred by several different ways. Probiotics can colonize the gut and occupy niches that would have been available to potential pathogens as reviewed (111). This is competitive exclusion. Probiotics can achieve this by using up nutritional resources, or altering the environment (pH, mucus production) to make conditions undesirable for other microbes (112). Beneficial bacteria can also alter gene expression of potential pathogens so that virulence decreases (113). These microbes work synergistically with the intestinal community to maintain health. Many of the genera used, such as Lactobacillus, are predominant members of the intestinal community, and functionally important. Therefore, these bacteria colonize and persist in the host long enough to convey benefits. The mechanism of action of many probiotics is enhanced abilities to implement functions that are done by the resident microbiota. Probiotics can act through modulation of the host responses. They may induce the production of defensins or produce proteases 30 that cleave and activate this antimicrobial peptide (114). Bacteroides thetaiotaomicron, a resident microbe in the human gut, also has this effect (11). Probiotics can also activate expression of immuno-regulatory genes. Lactobacillus rhamnosus GG activate NF-κB in macrophages (115). They activate the immune response and allow hosts to clear potential pathogens. Conversely, another species, Lactobacillus plantarum, act by blocking signaling to this gene and suppresses inflammation (116). It was found that solution from L. plantarum culture inhibited the NF-κB expression of murine intestinal epithelial cells. They found that L. plantarum medium (without bacterium) inhibited NF-κB activation from MyD88 dependent and independent pathways via TNF receptor. Sometimes it is the bacterial metabolites, and not direct immunological effects of the microbes themselves that have health benefits. Martin et al. studied effects of two probiotics, Lactobacillus paracasei and L. rhamnosus in mice colonized with human baby flora (HBF) (117). Presence of these bacteria altered metabolitic profiles from blood and tissue of mice. L. paracasei changed energy metabolism pathways of the host by allowing accumulation of bile acids (bacteria cannot deconjugate this compound). This led to high lipid absorption in the intestine. Efficient metabolism of soybean oil from mouse chow also allowed mice to reduce plasma lipoprotein and liver cholesterol. Bacteria also changed metabolism of amino acids via glucogenesis. Wostmann summarizes studies done on contributions of the microbiota on biosynthetic pathways in the gut (13). Bacteria change lipid composition of fecal content and vitamin and mineral uptake in the gut. Also, complex carbohydrates are broken down into short chain fatty acids 31 with the aid of gut microbes. These functions indicate that it is a product of bacterium and not the microbe itself that has beneficial effects exemplifying various ways microorganisms affects the community. Conclusion Mammalian microbota is, without a doubt, a complex and dynamic ecosystem. Not only do microbes interact with each other to exist harmoniously (among resident microbes) or antagonistically (attack by parasites or predators such as Bdellovibrio), but also communicate and cooperate with the host. The gut microbiota is important in many aspects of host health. From discussions above, it can be seen that it is necessary for the microbiota to be resistant and resilient to disruptive forces in order for its function to be maintained. In the event that diversity of the gut community is altered, diseases such as IBD may develop. To study the relationship between the microbiota, host and disease, I intended to use known models of IBD to induce disease in mice and investigate subsequent changes in the intestinal microbiota. Studies have been done associating changes in the microbiota with disease, but none investigating changes in the DSS model in mice, or whether the relative abundance of H. hepaticus in the gut of IL-10-/- mice impact on disease severity. I intended to address these questions through experiments conducted in the upcoming chapters in order to attempt to identify possible roles of the microbiota in influencing disease. 32 REFERENCES 33 REFERENCES 1. Bäckhed F, Ley R, Sonnenburg J, et al. Host-bacterial mutualism in the human intestine. Science. 2005;307:1915-1920 2. Gill SR, Pop M, DeBoy RT, et al. Metagenomic Analysis of the Human Distal Gut Microbiome. Science. 2006;312:1355-1359 3. Dunbar J, Barns S, Ticknor L, et al. Empirical and theoretical bacterial diversity in four Arizona soils. Appl Environ Microbiol. 2002;68:3035-3045 4. Venter J, Remington K, Heidelberg J, et al. Environmental genome shotgun sequencing of the Sargasso Sea. Science. 2004;304:66-74 5. Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the Human Intestinal Microbial Flora. Science. 2005;308:1635-1638 6. Ley RE, Peterson DA, Gordon JI. Ecological and Evolutionary Forces Shaping Microbial Diversity in the Human Intestine. Cell. 2006;24:837-848 7. Chang JY, Antonopoulos DA, Kalra A, et al. Decreased Diversity of the Fecal Microbiome in Recurrent Clostridium difficile-Associated Diarrhea. J Infect Dis. 2008;197:435 8. Lepage P, Seksik P, Sutren M, et al. Biodiversity of the mucosaassociated microbiota is stable along the distal digestive tract in healthy individuals and patients with IBD. Inflamm Bowel Dis. 2005;11:473-480 9. Dethlefsen L, McFall-Ngai M. & Relman, DA. An ecological and evolutionary perspective on human–microbe mutualism and disease. Nature. 2007;449:811-818 10. Young VB, Schimdt TM. Antibiotic-Associated Diarrhea Accompanied by Large-Scale Alterations in the Composition of the Fecal Microbiota. J Clin Microbiol. 2004;42:1203-1206 34 11. Stappenbeck T, Hooper L, Gordon J. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. PNAS. 2002;99:15451-15455 12. Cummings JH, Macfarlane GT. Role of intestinal bacteria in nutrient metabolism. JPEN J Parenter Enteral Nutr. 1997;21:357-365 13. Wostmann B. The germfree animal in nutritional studies. Ann Rev Nutr. 1981;1 14. Wostmann B, Bruckner-Kardoss E. Development of cecal distention in germ-free baby rats. Am J Physiol. 1959;197 15. Gordon H, Bruckner-Kardoss E. Effect of the normal microbial flora on various tissue elements of the small intestine. Acta Anat (Basel). 1961;44 16. Levenson S. The influence of the indigenous microflora on mammalian metabolism and nutrition. JPEN J Parenter Enteral Nutr 1978;2:78-107 17. Ahmad MS, Krishnan S, Ramakrishna BS, et al. Butyrate and glucose metabolism by colonocytes in experimental colitis in mice. Gut. 2000;46:493-499 18. Hill R, Cowley H. The influence of colonizing micro-organisms on development of crypt architecture in the neonatal mouse colon. Acta Anat (Basel). 1990;137:137-140 19. Ukena S, Singh A, Dringenberg U, et al. Probiotic Escherichia coli Nissle 1917 inhibits leaky gut by enhancing mucosal integrity. PLoS One. 2007;2:e1308 20. Round J, Mazmanian S. The gut microbiota shapes intestinal immune responses during health and disease. Nature Rev Immunol. 2009;9:313323 21. Brandl K, Plitas G, Mihu C, et al. Vancomycin-resistant enterococci exploit antibiotic-induced innate immune deficits. Nature. 2008;455:804-807 35 22. Torruellas J, Jackson M, Pennock J, et al. The Yersinia pestis type III secretion needle plays a role in the regulation of Yop secretion. Mol Microbiol. 2005;57:1719-1733 23. Stecher B, Hapfelmeier S, Müller C, et al. Flagella and chemotaxis are required for efficient induction of Salmonella enterica serovar Typhimurium colitis in streptomycin-pretreated mice. Infect Immun. 2004;72:4138-4150 24. Kelly D, Campbell J, King T, et al. Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-gamma and RelA. Nature Immunol. 2004;5:104-112 25. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, et al. Recognition of Commensal Microflora by Toll-Like Receptors Is Required for Intestinal Homeostasis. Cell. 2004;118:229-241 26. Cario E, Gerken G, Podolsky DK. Toll-Like Receptor 2 Controls Mucosal Inflammation by Regulating Epithelial Barrier Function. Gastroenterol. 2007;132:1359 27. Cario E, Podolsky D. Intestinal epithelial TOLLerance inTOLLerance of commensals. Mol Immunol. 2005;42:887-893 28. Hu D, Zhu G, Mori F, et al. Staphylococcal enterotoxin induces emesis through increasing serotonin release in intestine and it is downregulated by cannabinoid receptor 1. Cell Microbiol. 2007;9:2267-2277 29. Sudo N, Chida Y, Aiba Y, et al. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 2004;558:263-275 30. Amaral F, Sachs D, Costa V, et al. Commensal microbiota is fundamental for the development of inflammatory pain. PNAS. 2008;105:2193-2197 31. Dumas M, Barton R, Toye A, et al. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. PNAS. 2006;103:12511-12516 36 versus 32. Ley RE, Backhed F, Turnbaugh P, et al. Obesity alters gut microbial ecology. PNAS. 2005;102:11070-11075 33. Orešič M, Seppänen-Laakso T, Yetukuri L, et al. Gut microbiota affects lens and retinal lipid composition. Experimental Eye Research 2009;89:604-607 34. Staley J, Konopka A. Measurement of in situ activities of nonphotosynthetic microorganisms in aquatic and terrestrial habitats. Ann Rev Microbiol. 2006;39:321-346 35. Woese C, Kandler O, Wheelis M. Towards a natural system of organisms: proposal for the domains Archaea, Bacteria, and Eucarya. PNAS. 1990;87:4576-4579 36. Ashelford KE, Chuzhanova NA, Fry JC, et al. At Least 1 in 20 16S rRNA Sequence Records Currently Held in Public Repositories Is Estimated To Contain Substantial Anomalies. Appl Environ Microbiol. 2005;71:77247736 37. Muyzer G, de Waal E, Uitterlinden A. Profiling of complex microbial populations by denaturing gradient gel electrophoresis analysis of polymerase chain reaction-amplified genes coding for 16S rRNA. Appl Environ Microbiol. 1993;59:695-700 38. Uenishi G, Fujita S, Ohashi G, et al. Molecular analyses of the intestinal microbiota of chimpanzees in the wild and in captivity. Am J Primatol. 2007;69:367-376 39. Keuhl CJ, Wood HD, Marsh TL, et al. Colonization of the Cecal Mucosa by Helicobacter hepaticus Impacts the Diversity of the Indigenous Microbiota. Infect Immun. 2005;73:6952-6961 40. Franks AH, Harmsen HJM, Raangs GC, et al. Variations of Bacterial Populations in Human Feces Measured by Fluorescent In Situ Hybridization with Group-Specific 16S rRNA-Targeted Oligonucleotide Probes. Appl Environ Microbiol. 1998;64:3336-3345 37 41. Salzman NH, de Jong H, Paterson Y, et al. Analysis of 16S libraries of mouse gastrointestinal microflora reveals a large new group of mouse intestinal bacteria. Microbiol. 2002;148:3651-3660 42. Ge Z, White D, Whary M, et al. Fluorogenic PCR-Based Quantitative Detection of a Murine Pathogen, Helicobacter hepaticus. J Clin Microbiol. 2001 39:2598-2602 43. Chan V, Crocetti G, Grehan M, et al. Visualization of Helicobacter species within the murine cecal mucosa using specific fluorescence in situ hybridization. Helicobacter. 2005;10:114-124 44. Temperton B, Field D, Oliver A, et al. Bias in assessments of marine microbial biodiversity in fosmid libraries as evaluated by pyrosequencing. ISME J. 2009;3:792-796 45. Neufeld J, Li J, Mohn W. Scratching the surface of the rare biosphere with ribosomal sequence tag primers. FEMS Microbiol Lett. 2008;283:146-153 46. Antonopoulos D, Huse S, Morrison H, et al. Reproducible community dynamics of the gastrointestinal microbiota following antibiotic perturbation. Infect Immun. 2009;77:2367-2375 47. Margulies M, Egholm M, Altman W, et al. Genome sequencing in microfabricated high-density picolitre reactors. Nature 2005;437:376-380 48. Ronaghi M, Uhlén M, Nyrén P. A sequencing method based on real-time pyrophosphate. Science. 1998;281:363-365 49. Dethlefsen L, Huse S, Sogin M, et al. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008;6:e280 50. Zoetendal E, von Wright A, Vilpponen-Salmela T, et al. Mucosaassociated bacteria in the human gastrointestinal tract are uniformly distributed along the colon and differ from the community recovered from feces. Appl Environ Microbiol. 2002;68:3401-3407 38 51. Garner C, Antonopoulos D, Wagner B, et al. Perturbation of the small intestine microbial ecology by streptomycin alters pathology in a Salmonella enterica serovar typhimurium murine model of infection. Infect Immun. 2009;77:2691-2702 52. Allison S, Martiny J. Colloquium paper: resistance, resilience, and redundancy in microbial communities. PNAS. 2008;105:11512-11519 53. Swidsinski A, Ladhoff A, Pernthaler A, et al. Mucosal flora in inflammatory bowel disease. Gastroenterol. 2002;122:44-54 54. McCann. K. The diversity-stability debate. Nature. 2000;405:228-233 55. Odum E. Fundamentals of ecology. 1953 56. Elton C. Ecology of invasions by Animals and Plants. 1958 57. Tilman D, Lehman C, Bristow C. Diversity-stability relationships: statistical inevitability or ecological consequence? Am Nat. 1998;151:277-282 58. MacArthur RH. Fluctuations of animal populations and measure of community stability. Ecology. 1955;36:533-536 59. May RM. Stability and complexity in model systems. 1973 60. Naeem S. Species redundancy and ecosystem reliability. Conservation Biology 1998;12:39-45 61. Naeem S. Biodiversity: 2002;416:23-24 62. Pfisterer A, Schmid B. Diversity-dependent production can decrease the stability of ecosystem functioning. Nature. 2002;416:84-86 63. Pryde SE, Duncan SH, Hold GL, et al. The microbiology of butyrate formation in the human colon. FEMS Microbiol Lett. 2002;217:133-139 biodiversity 39 equals instability? Nature. 64. Ley R, Lozupone C, Hamady M, et al. Worlds within worlds: evolution of the vertebrate gut microbiota. Nat Rev Microbiol. 2008;6:776-788 65. Biasucci G, Benenati B, Morelli L, et al. Cesarean delivery may affect the early biodiversity of intestinal bacteria. J Nutr. 2008;138:1796S-1800S 66. Nakamura N, Gaskins H, Collier C, et al. Molecular ecological analysis of fecal bacterial populations from term infants fed formula supplemented with selected blends of prebiotics. Appl Environ Microbiol. 2009;75:11211128 67. Penders J, Thijs C, Vink C, et al. Factors influencing the composition of the intestinal microbiota in early infancy. Pediatrics. 2006;118:511-521 68. Palmer C, Bik E, DiGiulio D, et al. Development of the human infant intestinal microbiota. PLoS Biol. 2007;5:e177 69. Ouwehand A, Salminen S, Arvola T, et al. Microbiota composition of the intestinal mucosa: association with fecal microbiota? Microbiol Immunol. 2004;48:497-500 70. Sakata S, Tonooka T, Ishizeki S, et al. Culture-independent analysis of fecal microbiota in infants, with special reference to Bifidobacterium species. FEMS Microbiol Lett. 2005;243:417-423 71. Enck P, Zimmermann K, Rusch K, et al. The effects of ageing on the colonic bacterial microflora in adults. Z Gastroenterol. 2009;47:653-658 72. Hayashi H, Sakamoto M, Benno Y. Fecal microbial diversity in a strict vegetarian as determined by molecular analysis and cultivation. Microbiol Immunol. 2002;46:819-831 73. Mueller S, Saunier K, Hanisch C, et al. Differences in fecal microbiota in different European study populations in relation to age, gender, and country: a cross-sectional study. Appl Environ Microbiol. 2006;72:10271033 74. Batt R, Rutgers H, Sancak A. Enteric bacteria: friend or foe? J Small Anim Pract. 1996;37:261-267 40 75. Wang M, Ahrne S, Jeppsson B, et al. Comparison of bacterial diversity along the human intestinal tract by direct cloning and sequencing of 16S rRNA genes. Volume 54: FEMS Microbiol Ecol. 2005:219-231 76. Lundin A, Bok CM, Aronsson L, et al. Toll-like receptors and nuclear receptors: a tripartite communication that tunes innate immunity in large intestine. Cell Microbiol. 2008;10:1093-1103 77. Snipes R. Anatomy of the cecum of the laboratory mouse and rat. Anat Embryol. 1981;162:455-474 78. Suzuki H. Differences in Intraepithelial Lymphocytes in the Proximal, Middle, Distal Parts of Small Intestine, Cecum, and Colon of Mice. Immunol Inves. 2009;38:780-796 79. Takahashi N, Yamada T. Acid-induced acid tolerance and acidogenicity of non-mutans streptococci. Oral Microbiol Immunol. 1999;14:43-48 80. Wang X, Heazlewood S, Krause D, et al. Molecular characterization of the microbial species that colonize human ileal and colonic mucosa by using 16S rDNA sequence analysis. J Appl Microbiol. 2003;95:508-520 81. Swidsinski A, Loening-Baucke V, Vaneechoutte M, et al. Active Crohn's disease and ulcerative colitis can be specifically diagnosed and monitored based on the biostructure of the fecal flora. Inflamm Bowel Dis. 2008;14:147-161 82. Marteyn B, West NP, Browning DF, et al. Modulation of Shigella virulence in response to available oxygen in vivo. Nature 2010;465:355-358 83. Suau A, Bonnet R, Sutren M, et al. Direct Analysis of Genes Encoding 16S rRNA from Complex Communities Reveals Many Novel Molecular Species within the Human Gut. Appl Environ Microbiol. 1999;65:47994807 84. Finegold S, Sutter V, Sugihara P, et al. Fecal microbial flora in Seventh Day Adventist populations and control subjects. Am J Clin Nutri. 1977;30:1781-1792 41 85. Kingston D, Van Tassell R, Wilkins T. The fecapentaenes, potent mutagens from human feces. Chem Res Toxicol. 1990;3:391-400 86. Barman M, Unold D, Shifley K, et al. Enteric salmonellosis disrupts the microbial ecology of the murine gastrointestinal tract. Infect Immun. 2008;76:907-915 87. Croswell A, Amir E, Teggatz P, et al. Prolonged impact of antibiotics on intestinal microbial ecology and susceptibility to enteric Salmonella infection. Infect Immun. 2009;77:2741-2753 88. Van Der Waaij D, Strum C. Antibiotic Decontamination of the Digestive Tract of Mice. Technical Procedures. Lab Anim Care. 1968;18:1-10 89. Thoreson R, Cullen J. Pathophysiology of inflammatory bowel disease: an overview. Surg Clin North Am. 2007;87:575-585 90. Rath H, Schultz M, Freitag R, et al. Different Subsets of Enteric Bacteria Induce and Perpetuate Experimental Colitis in Rats and MIce. Infect Immun. 2001;69:2277-2285 91. Rath HC, Ikeda JS, Linde HJ, et al. Varying cecal bacterial loads influences colitis and gastritis in HLA-B27 transgenic rats. Gastroenterol. 1999;116:310-319 92. Sellon R, Tonkonogy S, Schultz M, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66:52245231 93. Sydora B, Martin S, Lupicki M, et al. Bacterial antigens alone can influence intestinal barrier integrity, but live bacteria are required for initiation of intestinal inflammation and injury. Inflamm Bowel Dis. 2006;12:429-436 94. Ohkusa T, Yoshida T, Sato N, et al. Commensal bacteria can enter colonic epithelial cells and induce proinflammatory cytokine secretion: a possible pathogenic mechanism of ulcerative colitis. J Med Microbiol. 2009;58:535-545 42 95. Young VB, Knox KA, Pratt JS, et al. In Vitro and In Vivo Characterization of Helicobacter hepaticus Cytolethal Distending Toxin Mutants. Infec Immun. 2004;72:2521-2527 96. Okayasu I, Hatakeyama S, Yamada M, et al. A Novel Method in the Induction of reliable Experimental Acute and Chronic Ulcerative Colitis in Mice. Gastroenterol. 1990;98:694-702 97. Kitajima S, Takuma S, Morimoto M. Changes in Colonic Mucosal Permeability in Mouse Colitis Induced with Dextran Sulfate Sodium. Exper Anim. 1999;48:137-143 98. Wirtz S, Neufert C, Weigmann B, et al. Chemically induced mouse models of intestinal inflammation. Nature Protoc. 2007;2:541-546 99. Kitajima S, Morimoto M, Sagara E. A Model of Dextran Sodium Supfateinduced Mouse Colitis: Bacterial Degradation of DSS Does Not Occur after Incubation with Mouse Cecal Contents. Exper Anim. 2002;51:203206 100. Ito R, Kita M, Shin-Ya M, et al. Involvement of IL-17A in the pathogenesis of DSS-induced colitis in mice. Biochem Biophys Res Commun. 2008;377:12-16 101. Yan Y, Kolachala V, Dalmasso G, et al. Temporal and spatial analysis of clinical and molecular parameters in dextran sodium sulfate induced colitis. PLoS One 2009;4:e6073 102. Rakoff-Nahoum S, Hao L, Medzhitov R. Role of Toll-like Receptors in Spontaneous Commensal-Dependent Colitis. Immun. 2006;25:319-329 103. Ungaro R, Fukata M, Hsu D, et al. A novel Toll-like receptor 4 antagonist antibody ameliorates inflammation but impairs mucosal healing in murine colitis. Am J Physiol, Gastroint Liver Physiol. 2009;296:G1167-79 104. Cahill R, Foltz C, Fox J, et al. Inflammatory bowel disease: an immunitymediated condition triggered by bacterial infection with Helicobacter hepaticus. Infect Immun. 1997;65:3126-3131 43 105. Pratt JS, Sachen KL, Wood HD, et al. Modulation of host immune responses by the Cytolethal Distending Toxin of Helicobacter hepaticus. Infect Immun. 2006;74:4406-4503 106. Ge Z, Sterzenbach T, Whary M, et al. Helicobacter hepaticus HHGI1 is a pathogenicity island associated with typhlocolitis in B6.129-IL10 tm1Cgn mice. Microbes Infect. 2008;10:726-733 107. Kullberg M, Jankovic D, Feng C, et al. IL-23 plays a key role in Helicobacter hepaticus-induced T cell-dependent colitis. J Exp Med. 2006;203:2485-2494 108. Dieleman L, Arends A, Tonkonogy S, et al. Helicobacter hepaticus does not induce or potentiate colitis in interleukin-10-deficient mice. Infect Immun. 2000;68:5107-13 109. Peran L, Camuesco D, Comalada M, et al. Lactobacillus fermentus, a probiotic capable to release glutothione, prevents colonic inflammation in the TNBS model of rat colitis. Int J Colorectal Dis. 2006;21:737-746 110. Herias MV, Koninkx JF, Vos JG et al. Probiotic effects of Lactobacillus casei on DSS-induced ulcerative colitis in mice. Int J Food Microbiol. 2005;103:143-155 111. Callaway T, Edrington T, Anderson R, et al. Probiotics, prebiotics and competitive exclusion for prophylaxis against bacterial disease. Anim Health Res Rev. 2008;9:217-225 112. Spurbeck R, Arvidson C. Inhibition of Neisseria gonorrhoeae epithelial cell interactions by vaginal Lactobacillus species. Infect Immun. 2008;76:31243130 113. Medellin-Peña M, Wang H, Johnson R, et al. Probiotics affect virulencerelated gene expression in Escherichia coli O157:H7. Appl Environ Microbiol. 2007;73:4259-4267 114. Wilson C, Ouellette A, Satchell D, et al. Regulation of intestinal alphadefensin activation by the metalloproteinase matrilysin in innate host defense. Science. 1999;286:113-117 44 115. Miettinen M, Lehtonen A, Julkunen I, et al. Lactobacilli and Streptococci activate NF-kappa B and STAT signaling pathways in human macrophages. J Immunol. 2000;164:3733-3740 116. Petrof E, Claud E, Sun J, et al. Bacteria-free solution derived from lactobacillus plantarum inhibits multiple NF-kappaB pathways and inhibits proteasome function. Inflamm Bowel Dis. 2009;15:1537-1547 117. Martin F-PJ, Dumas M-E, Wang Y, et al. A top-down systems biology view of microbiome-mammalian metabolic interactions in a mouse model. Mol Syst Biol. 2007;3 45 Chapter 2 Models of IBD There are many models of inflammatory bowel disease (IBD) geared towards understanding various aspects of this idiopathic disease. They address issues that are difficult to investigate in humans, such as changes in early stages of disease and effects of new therapies. Generally, these models are categorized into three groups: chemical, immune dysregulation and environmental. However these groups often overlap and some models involve more than just one factor, for example in the case of immuno-compromised animals exposed to antigens employ both immunological and environmental triggers (1, 2). These environmental triggers may include luminal stimulation from ingested material as well as from the microbial community present in the gut. The modern view on the role of luminal microbial communities extends from invasion of pathogens, to alterations in resident community structure, also known as dysbiosis. However, changes in the microbiota associated with disease in many of these models are yet to be characterized. Moreover, roles of the microbiota at various stages of disease, initiation and progression, as well as exacerbation or reduction of inflammation, are still to be determined. Here, we choose two well-known models of IBD, DSS-induced colitis and the Helicobacter hepaticus triggered IL-10 deficient murine model, to investigate 46 various roles of the microbiota in IBD. The former causes disease in mice that resembles human colitis, where inflammation is superficial, occurring in the mucosa and submucosa, as well as being confined to the lower intestine. Although there is some evidence that the microbiota plays a role in stimulating immune dysregulation in this model, there has been limited evidence that alterations in community structure occur after DSS administration, and no studies characterizing changes that may occur over the course of disease progression. The latter resembles Crohn’s disease in humans due to its Th1 immune response and non-contiguous pattern of mucosal inflammation that occurs in deep layers of the intestine. Importance of the role of the microbiota is inherent in this model since a microbe invades the resident community and triggers disease. However, the role of the microbiota in disease progression is unknown. Before presenting experiments designed to address some of these questions at hand, the following chapter aims to describe models that will be used in upcoming studies. Value of animal models Animal models provide a means to test questions that may be difficult or impossible to investigate in humans. An ideal model should be inexpensive, easy to use and reproducible. There are typically several models designed to study a single disease due to many aspects of physiology, immunology, microbiology and genetics that interact to maintain a healthy biological system. A model is 47 chosen since it mimics, or is believed to mimic, a feature of the disease as is it is occurs in humans. Often times the condition studied in an animal model may not represent human disease entirely, but rather only certain aspects are recapitulated, hence the use of several models to answer questions about one disease. Each model is typically used to answer specific questions and can be investigated at various phase of disease where pathogenic components may be scrutinized. Some conditions, such as IBD, are very complex and occur by interplay of several mechanisms making prognosis and treatment challenging. As such, several models have been developed to study disease. These systems have proven to be very important in teasing apart various mechanisms by which disease may be caused (3, 4). For example, models utilizing TLR, IL-10 and IL-2 deficient mice have been used to investigate various roles of the immune system and genetic defects in disease (5-7). Furthermore, the role of the microbiota has been studied using germfree and antibiotic treated animal models (5, 6); and chemically induced disease models have been used to study loss of mucosal tolerance and break-down of the epithelial barrier as potential mechanisms of disease development (7, 8). Two-widely used animal models in IBD There are several types of animal models available to study various aspects of IBD (3). They are generally divided into four groups: Those that are triggered by 48 dysfunction of innate immunity such as A20 deficient mice (9), by dysfunction of adaptive immunity such as CD45RBHi transfer models (10), by the environment such as H. hepaticus triggered disease in immuno-compromised mice and by chemicals such as DSS and TNBD- induced disease (1, 8, 11). Here we focus on two models investigated in following chapters: DSS-induced colitis and H. hepaticus triggered disease in IL-10 deficient mice. DSS-induced colitis This murine model has been used since 1990 when Okayasu et al. modified it from the original guinea pig system (8). Disease produced in this model is highly dependent on mouse strain used as well as concentration of DSS administered and duration of treatment (12-14). Variations of this model can also lead to acute or chronic disease, and carcinomas (15). DSS-induced colitis in mice can progress to dysplasia then carcinoma, similar to the disease sequence in humans. This model of disease also responds to therapies administered to humans such as steroids (16) and antibiotics (16), and has an immunological response and genetic component (12). The model mimics aspects of human disease, and its outcome is reproducible. Origin of the model In 1961, the bacteria Leuconostoc spp. and Streptococcus spp. were found to produce a chemical polysaccharide called dextran from sucrose. Dextran sodium 49 sulfate (DSS) is produced by esterification of dextran with chlorosulfonic acid, which produces a molecule with 17% sulfur. This chemical has a large range in molecular weight, from 5000 to 1.4 million Da; however, usually 40 kDa is used in inducing colitis (13). Eight years after the discovery of DSS, the carrageenan model of IBD was developed when rodents fed with seaweed developed disease which led to the discovery of this sulfated polysaccharide that induced colitis (17). Later, due to its similar characteristics to carrageenan, DSS was also used to induce disease in hamsters (18), and then Okayasu and colleagues modified the model for mice (8). DSS has been administered to hamsters, rabbits, rats and mice, with larger rodents initially used because clinical signs could be easily monitored. Okayasu et al. found that mice administered 3 to 10% DSS solutions quickly developed inflammation resembling colitis in humans. They also found that changing the treatment regimen by cycling the DSS treatment with periods of water, instead of with continuous DSS treatment only, could induce chronic disease. Mechanism of the model The mechanism by which DSS-induced colitis develops is still unclear. However, it is generally believed that inflammation is triggered after the epithelial barrier is compromised by DSS, subsequently facilitating interactions between the host and resident microbes, which then triggers a host immune response that develops into inflammatory bowel disease. 50 DSS is thought to work by damaging the epithelial layers followed by immunological stimulation by resident microbes (19). Researchers found that DSS treatment led to reduction of zona occludens-1 protein production that is involved in maintenance of tight junctions and these changes preceded inflammation. There was also deposition of DSS in organs such as the liver and uptake by macrophages, adding to evidence of DSS permeability. Others have shown that DSS penetrates and disrupts the normally impervious mucin layer allowing bacteria to penetrate the barrier within 12 hours post-DSS administration, before inflammatory changes are detected (20). Another proposed mechanism for DSS action is by direct damage to intestinal cells. Investigators found that the chemical caused cytotoxicity that increased with time and concentration of DSS used (21). This group also found that expression of various cell adhesion molecules were affected by DSS, indicating that this alteration, particularly in β7 integrins, may lead to inflammation (21). Araki et al. found that DSS inhibits generation of reactive oxygen species (ROS) in Caco-2 cells, yet DSS administration in this murine model increases ROS, leading authors to suggest that ROS production comes from deeper cells and not the epithelial layer (22). DSS also increases permeability of the epithelial layer, as seen in human IBD (19), with significant increase in Evans Blue dye from the gut lumen after only 3 days of DSS administration, although no signs of inflammation or changes in architecture were seen at this time point (19). This group also showed that DSS may be responsible for basal crypt cell damage, and together with other cytotoxic 51 effects of DSS, it is clear that this chemical is involved in epithelial cell damage (21, 23, 24), an outcome that may play an important role in induction of disease. Another line of research suggests that the microbiota plays a major role in DSSinduced disease. Some have shown that germfree mice administered DSS develop more severe disease than their conventionally raised counterparts, suggesting a protective effect of the microbes (25). This indicates that the microbiota’s beneficial effects may be due to production of metabolites such as short chain fatty acids (SCFA) that are necessary for colonocyte development and epithelial cell health (26). However, others have shown that administration of anaerobic bacterial antigens reduced severity of disease in mice treated with DSS (27), leading these authors to speculate that ingested antigens increased tolerance to intestinal bacteria. This indicates that altered homeostasis between the microbiota and host immune response may be the driver for immune reaction to DSS (27). These effects may be due to the antigens altering the microbial community by competing for binding sites on epithelial surfaces, altering the immune system that may have subsequently exerted effects of the microbiota. Antibiotic studies show that certain antimicrobial therapies reduce inflammation (16), indicating both a protective and detrimental effect of the microbiota on colitis. Araki et al. treated Caco-2 cells in vitro with DSS and showed that H2S (22), thought to be associated with DSS-induced colitis in mice and UC in humans, was not produced by host reaction to the chemical, indicating that it may be a by-product of metabolism by the microbiota (28). Others have tried to explain the negative role of the microbiota by postulating that microbes act on 52 DSS and metabolize it to a form that causes inflammation. Kitajima et al. incubated DSS with luminal contents for 24 hours before the components were studied though spectrometry (29). They found that microbes did not metabolize DSS, dispelling this potential mechanism of action. However, other evidence still indicates an important role for the microbiota in this model of IBD (14, 20, 30). Although there are many unknowns surrounding the mechanism of DSS disease induction in mice, certain aspects of the model are known to be important. For example molecular size of DSS and strain of mouse utilized can affect disease severity. The molecular weight of the DSS administered to animals is relevant to disease induction also (13). Investigators found that after testing the effects of 5% DSS solution using three different weights of DSS, mice fed with DSS weights less than 5 kDa or over 500 kDa do not produce severe inflammation while 40 kDa is typically used to trigger colitis (13). Others have also found that the sulfur content was dose dependent on the ability of DSS to induce disease with amounts lower than 16.6 % causing a significant reduction in inducing inflammation (31). Authors speculated that larger molecular weights contained more sulfur that was important in inducing disease when molecules permeate the mucin layer, however DSS over 500 kDa are thought to be too large to penetrate this layer, thus unable to induce inflammation. This model is also dependent on strain of mice used with C57BL/6 mice being very susceptible to disease and BALB/c being more resistant (12). There are also strain dependent induction differences with BALB/c mice producing disease in the distal colon while Swiss Webster mice show more inflammation in colon and 53 cecum (32, 33). It is suggested that these differences are due to genetically controlled variation in inflammatory response or healing (34). Age and dose dependence was also seen in rats with 4 week old Wistar rats being more sensitive to DSS-induced colitis than those eight weeks old, and 4% DSS producing more severe inflammation that 2% DSS over 7 days administration (35). Immunology of the model Different phases of DSS-induced disease, acute and chronic, produce different inflammatory responses. The acute state is normally characterized by an increase in the Th1-Th17 response with up-regulation of TNFα, IL-6, IL-17 and KC (40). Other immunological characteristics of the acute state include up- regulation of IL-12, INFγ and minor roles of Th2 mediators IL-10 and IL-4 (36). A Th2 response is dominant in the chronic phase with a decrease in the aforementioned mediators and increase in IL-4 and IL-10 (37). Egger et al. also found that TNFα, IL-1 and IL-12 expression increased with increasing dose of DSS, but not volume of DSS solution ingested. Both Th1 and Th2 responses in Swiss Webster mice trigger DSS-induced colitis. Dieleman et al. showed that the acute phase of DSS colitis is initiated by neutrophils and macrophages and can lead to up-regulation of INFγ, IL-5 and IL4 expression (32), while the acute model requires macrophages predominantly while the chronic disease model requires both macrophages and T cells (23). 54 Leung et al. showed that gamma/delta T cells interact with LPS and macrophages before onset of inflammation and therefore likely to play an important role in DSS-induced colitis in rats (38). However, findings that SCID mice, which lack B and T cells, also developed disease after DSS treatment, indicate that macrophages are the main drivers (23). More recently, it was reported that the adaptive response is required for both acute and chronic phases of disease (39). Variation in the model This model was modified to be used in chronic colitis in hamsters in 1992 by Yamada et al. (40), before being used in mice. DSS causes disease in several strains of rodents with varying degrees of severity, allowing flexibility in choosing an appropriate system for the biologic question at hand (12, 15, 41). Guinea pigs develop severe inflammation within 3 days after start of DSS administration (42). Among mice, C3H/HeJ are highly susceptible to treatment with BALB/c mice being more resistant (12). Long-term administration of DSS results in colorectal carcinoma, very similar to dysplasia-carcinoma seen in cancer development in human ulcerative colitis (40, 43). Investigators treated hamsters with 1% DSS over 100 days and found that the animals developed colorectal cancer. This was to provide a model of disease seen in patients with long-term ulcerative colitis eventually developing colorectal carcinomas. 55 Another variation of the DSS model is also used to study dysplasia and carcinoma development by administering 3% of the chemical over seven days followed by water for 14 days, for three cycles (44). Or mice are given a single dose of azoxymethane (AOM) before three cycles of DSS prior to water treatment. Helicobacter hepaticus infected IL-10-/- murine model of IBD Helicobacter species are a relatively newly identified group of bacteria that has been documented to cause disease in humans and animals since the 1980’s (45). Diseases are varied and dependent on infecting bacterial species. For example, H. pylori causes peptic ulcers in humans, while H. rappini is associated with intestinal disease in human and other mammals (46), and abortions in guinea pigs (47). However, it was in 1992 that a new pathogen was found in mice at the National Cancer Institute-Frederick Cancer Research and Development Center (48), H. hepaticus, which was found in hepatic tissue of animals with liver tumors. It is also triggers inflammation in immuno-compromised mice that resembles human Crohn’s disease (1). This model relies on an immuno-altered background since H. hepaticus does not cause disease in wild-type mice (5, 49, 50). Disease resembles Crohn’s disease in humans with a Th1 response and transmural inflammation, but like many other models, it occurs in the lower intestine while human CD can occur anywhere along the gastrointestinal tract. This model is very desirable as it is highly 56 reproducible, easy and cheap to perform. This model is also conducted in SCID mice and severity of disease is dependent on mouse strains used (51). Origin of the model H. hepaticus is a microaerophilic, gram negative, flagellated (bipolar, sheathed), spiral-shaped bacterium (may be curved or have several spirals), 1.5 to 5.0 µm long and 0.2 to 0.3 µm wide. It has urease, oxidase and catalase activity, produces H2S, and reduces nitrate to nitrite (52). It is associated with the mucosal lining of the lower intestinal tract of mice and commonly occurs in the cecum and colon (53, 54). It is not known as a human pathogen; however, recent reports have indicated its presence in immuno-compromised children. At one point 88% of mice in laboratory and animal facilities were infected with H. hepaticus (55-57). This bacterium can be a normal part of the resident community of these mice and only in an immuno-altered state does it become pathogenic. In wild-type mice this bacterium can represent up to 40% of the microbiota (58). SCID mice and IL-10 deficient mice infected with H. hepaticus then became a useful model to study human IBD since it produces a Th1-Th17 type immune response similar to Crohn’s disease in humans. Mechanism of the model The genome of H. hepaticus has been completely characterized and has been shown to possess similarities to other pathogenic bacteria such as H. pylori 57 urease genes and the Campylobacter jejuni gene cluster that comprises cytolethal distending toxin (59). Although urease is not necessary for intestinal colonization, it plays a role in liver disease and triggers the immune response (60). Additionally, the HHGI1 region, a pathogenicity island, contains 70 coding regions which are mostly hypothetical proteins, three coding regions that are similar to regions that code for the type IV secretion system, that also plays a role in inducing disease. In a study with 12 strains of H. hepaticus it was observed that the HHGI1 region was either completely in some strains or partially detected in others which may be responsible for the varying virulence observed (29). H. hepaticus produces cytolethal distending toxin (Cdt), which not only arrests cell cycle in the G2 phase of cell division, but also has a role in colonization with the Cdt mutant being cleared from infected animals within eight months of infection (61). Researchers speculate that this may be due to the CdtB gene functioning to suppress host’s immune response, as seen by lower colonic inflammation in mice infected with the mutant, thereby allowing the bacterium to avoid detection or clearance (61). Disease does not occur in IL-10 deficient mice monoassociated with H. hepaticus indicating an important role of the resident microbiota in development of disease in this model of IBD (5). Furthermore, Helicobacter spp. have been found to cause dysregulation of TLR2 and NOD1 in epithelial cells (62), receptors for bacterial antigens. Investigation of the role of TLR4 and IL-10 in H. hepaticus triggered colitis show that TLR/IL-10 deficient mice have elevated INFγ, IL-17, TNFα, IL-1 and IL-6 when infected with this bacterium, emphasizing the role of 58 resident bacteria in induction of disease. Disease was more severe in double knock-out mice than in IL-10 deficient only mice, which may be related to increases the accumulation of apoptotic cells in the lamina propria in double knock-outs. Immunology of the model This model elicits a Th1/Th17 type response with the production of TNFα, IL-17 and IL-23 (63). Kullberg et al. found that both IL-23 and IL-12 have p40 subunits, which is an essential subunit in disease induction (63, 64). Previous work pointed to IL-12 as having an important role in H. hepaticus triggered disease in IL-10 deficient mice after the use of anti-p40 monoclonal antibodies, however these antibodies also targeted IL-23 (63). After researchers used more targeted approaches to investigate these two cytokines it was determined that IL-23 and not IL-12 is important in this model (63). Furthermore, it was found that in the absence of INFγ, IL-17 cannot produce severe disease. It was also found that disease was inhibited only in IL-23 deficient mice. Authors suggest that when IL23 is down-regulated by IL-10, lowered IL-23 levels then suppress IL-17 production from TH17 cells. This model hinges on the role of IL-10 in inhibiting diseases in mice. Wang et al. examined the role of NFκB on IL-10 by testing the role of subunits of NFκB in IL10 expression with the use of IL-10 immunoglobulin (Ig) fusion proteins in RAG mice (65). They found that the fusion protein prevented disease in H. hepaticus 59 infected RAG mice but not in mice deficient in p50. Radiation treatment further revealed that p50/p105 subunit of this transcription factor in homopoietic cells of the innate immune system is important in IL-10 expression and suppression of disease (66). This indicates that there is an important role for the innate immune system in H. hepaticus triggered colitis. Authors also found that c-Rel, a subunit of NfκB, is needed for H. hepaticus triggered inflammation. This subunit is necessary for proper functioning of IL-12/23 and absence prevented disease in RAG-2 deficient mice and prevented the ability of adoptive transfer of CD4+CD45RB high T cells to ameliorate disease (65). The model works via the adaptive response with inflammation occurring in SCID mice that had adoptive transfer of CD4+ T cells. Cahill et al. found that mice that were infected with H. hepaticus developed severe inflammation if they had CD4+ T cells expressing high levels of CD45RB (1). Disease did not develop in mice raised in germfree conditions indicating a role for resident bacteria, and a dysfunctional immune response, in disease development. Variations of this model There are many lines of mice that develop colitis when infected with H. hepaticus (51). SCID/NCr mice infected with H. hepaticus are used to demonstrate IBD. Natural infection with H. hepaticus resulted in lesions in the liver and intestines that progressed with age (49). Disease manifested was less severe than that observed in A/JCr mice (67). This indicates that the T cell response caused 60 reduced disease as also suggested by IgG antibody levels being lower in SCID mice compared to A/JCr mice. It is also possible that different microbiota present in these mice strain play a role in the different disease manifestations observed. Infection of H. hepaticus into BALB-Min and BALB-RagMin mice show that this bacterium is involved with tumors of the colon and not small intestine. These tumors are not dependent on severity of inflammation, but depend on organs on which they occur (68). Other species of Helicobacter have also been used in models of IBD, such as H. bilis, H. cindai and H. fennili. Severity of disease that develops varies with the species used. Co-infections of H. hepaticus and H. bilis produce colitis that later develops into dysplasia in P-Glycoprotein-deficient mdr1a-/- mice. H. hepaticus alone was found to delay colitis in these mice while mono-infection with H. bilis accelerated colitis development. The co-infection is thought to progress to dysplasia since the H. hepaticus slows progress of disease generated by the H. bilis by inducing regulatory T-cells, and allowing abnormal growth as cells are transformed by H. bilis activity (69). This system was proposed as a model to study dysplasia, hyperplasia and neoplasia in humans. Conclusions By using the DSS-induced model of colitis and the H. hepaticus triggered IBD model, roles of the microbiota in development of IBD can be assessed. Upcoming experiments attempt to investigate roles of the microbiota in initiation 61 and progression of disease. With the DSS-induced model of colitis, changes in the microbiota will be investigated at onset and progression of disease since there is no reported data documenting whether dysbiosis is associated with disease over time. Using the H. hepaticus triggered IBD model, where the microbiota is essential in onset of disease, the role of changes in the microbiota at initiation and during disease will also be investigated. By assessing how changes in the microbial community during development of IBD affect disease manifestation, advances towards prevention and therapy may be achieved. 62 REFERENCES 63 REFERENCES 1. Cahill R, Foltz C, Fox J, et al. Inflammatory bowel disease: an immunitymediated condition triggered by bacterial infection with Helicobacter hepaticus. Infect immun. 1997;65:3126-3131 2. Hermiston ML, Gordon JI. Inflammatory Bowel Disease and Adenomas in Mice Expressing a Dominant Negative N-Cadherin. Science. 1995;270:12031207 3. Wirtz S, Neurath MF. Mouse models of inflammatory bowel disease. Adv Drug Deliv Rev. 2007;59:1073-1083 4. Warren S. Why study animal models of IBD? Inflamm Bowel Dis. 2008;14:S129-S131 5. Dieleman L, Arends A, Tonkonogy S, et al. Helicobacter hepaticus does not induce or potentiate colitis in interleukin-10-deficient mice. Infection and Immunity. 2000;68:5107-5113 6. Madsen KL, Doyle JS, Tavernini MM, et al. Antibiotic therapy attenuates colitis in interleukin 10 gene-deficient mice. Gastroenterol. 2000;118:1094-1105 7. Wirtz S, Neufert C, Weigmann B, et al. Chemically induced mouse models of intestinal inflammation. Nat Protoc. 2007;2:541-546 8. Okayasu I, Hatakeyama S, Yamada M, et al. A Novel Method in the Induction of reliable Experimental Acute and Chronic Ulcerative Colitis in Mice. Gastroenterol. 1990;98:694-702 9. Hitotsumatsu O, Ahmad RC, Tavares R et al. The ubiquitin-editing enzyme A20 restricts nucleotide-binding oligomerization domain containing 2triggered signals. Immunity. 2008;28:381-390 10. Powrie F, Leach MW, Mauze S et al. Inhibition of Th1 responses prevents inflammatory bowel disease in scid mice reconstituted with CD45RBhi CD4+ T cells. Immunity. 1994;1:553-562 64 11. Scheiffele F, Fuss I. Induction of TNBS colitis in mice. Curr Protoc Immunol. 2002:15.19.11-15.19.14 12. Mahler M, Bristol JI, Leiter HE, et al. Differential susceptibility of inbred mouse strains to dextran sulphate sodium-induced colitis. Am J Physiol. 1998;274:544-551 13. Kitajima S, Takuma S, Morimoto M. Histological analysis of murine colitis induced by dextran sulfate sodium of different molecular weights. Exp Animals. 2000;49:9-15 14. Hudcovic T, Stepankova R, Cebra J, et al. The Role of Microflora in the Development of Intestinal Inflammation: Acute and Chronic Colitis Induced by Dextran Sulfate in Germ-Free and Conventionally Reared Immunocompetent and Immunodeficient Mice. Folia Microbiol (Praha). 2001;46:565-572 15. Suzuki R, Kohno H, Sugie S, et al. Strain differences in the susceptibility to azoxymethane and dextran sodium sulfate-induced colon carcinogenesis in mice. Carcinogenesis. 2006;27:162-169 16. Hans W, Schölmerich J, Gross V et al. The role of the resident intestinal flora in acute and chronic dextran sulfate sodium-induced colitis in mice. Eur J Gastroenterol Hepatol. 2000;12:267-273 17. Watt J, Marcus R. Carrageenan-induced ulceration of the large intestine in the guinea pig. Gut. 1971;12:164-171 18. Ohkusa T. Production of experimental ulcerative colitis in hamsters by dextran sulfate sodium and changes in intestinal microflora. Nippon Shokakibyo Gakkai Zasshi. 1985;82:1327-1336 19. Kitajima S, Takuma S, Morimoto M. Changes in Colonic Mucosal Permeability in Mouse Colitis Induced with Dextran Sulfate Sodium. Exp Anim. 1999;48:137-143 20. Johansson ME, Gustafsson JK, Sjöberg KE et al. Bacteria penetrate the inner mucus layer before inflammation in the dextran sulfate colitis model. PloS One. 2010;5:e12238 65 21. Ni J, Chen SF, Hollander D. Effects of dextran sulphate sodium on intestinal epithelial cells and intestinal lymphocytes. Gut. 1996;39:234-241 22. Araki Y, Sugihara H, Hattori T. In vitro effects of dextran sulfate sodium on a Caco-2 cell line and plausible mechanisms for dextran sulfate sodium-induced colitis. Oncol Rep. 2006;16:1357-1362 23. Dieleman LA, Ridwan BU, Tennyson GS et al. Dextran sulfate sodiuminduced colitis occurs in severe combined immunodeficient mice. Gastroenterol. 1994;107:1643-1652 24. Mashimo H, Wu D-C, Podolsky DK, et al. Impaired Defense of Intestinal Mucosa in Mice Lacking Intestinal Trefoil Factor. Science. 1996;274:262-265 25. Kitajima S, Morimoto M, Sagara E, et al. Dextran Sodium Sufate-Induced Colitis in Germ-Free IQI/Jic Mice. Exp Anim. 2001;50:387-395 26. Ahmad MS, Krishnan S, Ramakrishna BS, et al. Butyrate and glucose metabolism by colonocytes in experimental colitis in mice. Gut. 2000;46:493-499 27. Verdu EF, Bercik P, Cukrowska B, et al. Oral administration of antigens from intestinal flora anaerobic bacteria reduces the severity of experimemntal acute colitis in BALB/c mice. Clin Exp Immunol. 2000;120:46-50 28. Araki Y, Andoh A, Tsujikawa T et al. Alterations in intestinal microflora, faecal bile acids and short chain fatty acids in dextran sulphate sodium-induced experimental acute colitis in rats. Eur J Gastroenterol Hepatol. 2001;13:107-112 29. Belzer C, Stoof J, Breijer S, et al. The Helicobacter hepaticus hefA gene is involved in resistance to amoxicillin. Helicobacter. 2009;14:72-79 30. Rath H, Schultz M, Freitag R, et al. Different Subsets of Enteric Bacteria Induce and Perpetuate Experimental Colitis in Rats and MIce. Infect Immun. 2001;69:2277-2285 31. Axelsson LG, Landström E, Goldschmidt TJ et al. Dextran sulfate sodium (DSS) induced experimental colitis in immunodeficient mice: effects in CD4(+) cell depleted, athymic and NK-cell depleted SCID mice. Inflamm Res 1996;45:181-191 66 32. Dieleman LA, Palmen MJ, Akol H, et al. Chronic experimental colitis induced by dextran sulphate sodium (DSS) is characterized by Th1 and Th2 cytokines. Clin Expl Immunol. 1998;114:385-391 33. Dieleman LA, Ridwan BU, Tennyson GS et al. Dextran sulfate sodiuminduced colitis occurs in severe combined immunodeficient mice. Gastroenterol. 1994;107:1643-1652 34. Mähler M, Bristol IJ, Sundberg JP et al. Genetic analysis of susceptibility to dextran sulfate sodium-induced colitis in mice. Genomics. 1999;55:147-156 35. Shimizu T, Suzuki M, Fujimura J, et al. The Relationship Between the Concentration of Dextran Sodium Sulfate and the Degree of Induced Experimental Colitis in Weanling Rats. J Pediatr Gastroenterol Nutr. 2003;37:481-486 36. Egger B, Bajaj-Elliott M, MacDonald TT et al. Characterisation of acute murine dextran sodium sulphate colitis: cytokine profile and dose dependency. Digestion. 2000;62:240-248 37. Philip A, Nicholas CZ, Thuan N, et al. Distinct cytokine patterns identified from multiplex profiles of murine DSS and TNBS-induced colitis. Inflamm Bowel Dis. 2009;15:341-352 38. Leung FW, Heng M, Allen S et al. Involvement of luminal bacteria, heat shock protein 60, macrophages and gammadelta T cells in dextran sulfate sodium-induced colitis in rats. Dig Dis Sci. 2000;45:142-1479 39. Hall LJ, Faivre E, Quinlan A et al. Induction and Activation of Adaptive Immune Populations During Acute and Chronic Phases of a Murine Model of Experimental Colitis. Dig Dis Sci. 2010;Epub 40. Yamada M, Ohkusa T, Okayasu I. Occurrence of dysplasia and adenocarcinoma after experimental chronic ulcerative colitis in hamsters induced by dextran sulphate sodium. Gut. 1992;33:1521-1527 41. Stevceva L, Pavli P, Buffinton G, et al. Dextran sodium sulphate-induced colitis activity varies with mouse strain but develops in lipopolysaccharideunresponsive mice. J Gastroentorol Hepatol. 1999;14:54-60 67 42. Iwanaga T, Hoshi O, Han H et al. Morphological analysis of acute ulcerative colitis experimentally induced by dextran sulfate sodium in the guinea pig: some possible mechanisms of cecal ulceration. J Gastroenterol. 1994;29:430-438 43. Isao O, Masahiro Y, Tetuo M, et al. Dysplasia and carcinoma development in a repeated dextran sulfate sodium-induced colitis model. J Gastroenterol Hepatol. 2002;17:1078-1083 44. Okayasu I, Yamada M, Mikami T, et al. Dysplasia and carcinoma development in a repeated dextran sulfate sodium-induced colitis model. J Gastroenterol Hepatol. 2002;17:1078-1083 45. Marshall BJ, Warren JR. Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet. 1984;1:1311-1315 46. Schauer DB, Ghori N, Falkow S. Isolation and characterization of "Flexispira rappini" from laboratory mice. J Clin Microbiol. 1993;31:2709-2714 47. Bryner JH, Ritchie AE, Pollet L et al. Experimental infection and abortion of pregnant guinea pigs with a unique spirillum-like bacterium isolated from aborted ovine fetuses. Am J Vet Res. 1987;48:91-95 48. Ward JM, Fox JG, Anver MR, et al. Chronic Active Hepatitis and Associated Liver Tumors in Mice Caused by a Persistent Bacterial Infection With a Novel Helicobacter Species. J Natl Cancer Inst. 1994;86:1222-1227 49. Li X, Fox JG, Whary MT, et al. SCID/NCr Mice Naturally Infected with Helicobacter hepaticus Develop Progressive Hepatitis, Proliferative Typhlitis, and Colitis. Infect Immun. 1998;66:5477-5484 50. Chow J, Mazmanian S. A Pathobiont of the Microbiota Balances Host Colonization and Intestinal Inflammation. Cell Host Microbe. 2010;7:265-276 51. Foltz CJ, Fox JG, Cahill R, et al. Spontaneous Inflammatory Bowel Disease in Multiple Mutant Mouse Lines: Association with Colonization by Helicobacter hepaticus. Helicobacter. 1998;3:69-78 68 52. Fox JG, Dewhirst FE, Tully JG, et al. Helicobacter hepaticus sp. nov., a microaerophilic bacterium isolated from livers and intestinal mucosal scrapings from mice. J Clin Microbiol. 1994;32:1238-1245 53. Chan V, Crocetti G, Grehan M, et al. Visualization of Helicobacter species within the murine cecal mucosa using specific fluorescence in situ hybridization. Helicobacter. 2005;10:114-124 54. Zhang L, Danon SJ, Grehan M et al. Natural colonization with Helicobacter species and the development of inflammatory bowel disease in interleukin-10-deficient mice. Helicobacter. 2005;10:223-230 55. Bohr UR, Selgrad M, Ochmann C, et al. Prevalence and Spread of Enterohepatic Helicobacter Species in Mice Reared in a Specific-Pathogen-Free Animal Facility. J Clin Microbiol. 2006;44:738-742 56. Goto K, Ohashi H, Takakura A, et al. Current status of Helicobacter contamination of laboratory mice, rats, gerbils, and house musk shrews in Japan. Curr Microbiol. 2000;41:161-166 57. Nilsson HO, Ouis IS, Stenram U, et al. High Prevalence of Helicobacter Species Detected in Laboratory Mouse Strains by Multiplex PCR-Denaturing Gradient Gel Electrophoresis and Pyrosequencing. J Clin Microbiol. 2004;42:3781-3788 58. Keuhl CJ, Wood HD, Marsh TL, et al. Colonization of the Cecal Mucosa by Helicobacter hepaticus Impacts the Diversity of the Indigenous Microbiota. Infect Immun. 2005;73:6952-6961 59. Suerbaum S, Josenhans C, Sterzenbach T, et al. The complete genome sequence of the carcinogenic bacterium Helicobacter hepaticus. PNAS. 2003;100:7901-7906 60. Beckwith CS, McGee DJ, Mobley HL, et al. Cloning, Expression, and Catalytic Activity of Helicobacter hepaticus Urease. Infect Immun. 2001;69:59145920 61. Pratt JS, Sachen KL, Wood HD, et al. Modulation of host immune responses by the Cytolethal Distending Toxin of Helicobacter hepaticus. Infect Immun. 2006;74:4406-4503 69 62. Chaouche-Drider N, Kaparakis M, Karrar A, et al. A commensal Helicobacter sp. of the rodent intestinal flora activates TLR2 and NOD1 responses in epithelial cells. PLoS One. 2009;4:e5396 63. Kullberg M, Jankovic D, Feng C, et al. IL-23 plays a key role in Helicobacter hepaticus-induced T cell-dependent colitis. J Exp Med. 2006;203:2485-2494 64. Kullberg MC, Rothfuchs AG, Jankovic D, et al. Helicobacter hepaticusInduced Colitis in Interleukin-10-Deficient Mice: Cytokine Requirements for the Induction and Maintenance of Intestinal Inflammation. Infect Immun. 2001;69:4232-4241 65. Wang Y, Rickman BH, Poutahidis T, et al. c-Rel Is Essential for the Development of Innate and T Cell-Induced Colitis. J Immunol. 2008;180:81188125 66. Tomczak M, Erdman S, Davidson A, et al. Inhibition of Helicobacter hepaticus-induced colitis by IL-10 requires the p50/p105 subunit of NF-kappa B. J Immunol. 2006;177:7332-7339 67. Whary MT, Morgan TJ, Dangler CA, et al. Chronic Active Hepatitis Induced by Helicobacter hepaticus in the A/JCr Mouse Is Associated with a Th1 Cell-Mediated Immune Response. Infect Immun. 1998;66:3142-3148 68. Nagamine CM, Rogers AB, Fox JG, et al. Helicobacter hepaticus promotes azoxymethane-initiated colon tumorigenesis in BALB/c-IL10-deficient mice. Int J Cancer. 2008;122:832-838 69. Maggio-Price L, Bielefeldt-Ohmann H, Treuting P, et al. Dual Infection with Helicobacter bilis and Helicobacter hepaticus in P-Glycoprotein-Deficient mdr1a/- Mice Results in Colitis that Progresses to Dysplasia. Am J Pathol. 2005;166:1793-1806 70 This chapter has been published: Nagalingam NA, Kao JK, Young VB. Microbial ecology of the murine gut associated with the development of dextran sodium sulphate-induced colitis. Inflamm Bowel Dis. 2010 I performed experiments, data retrieval and analyses. Dr. Kao provided custom primers for detection of changes in expression of host mediators, scored histology and provided discussion on observed host changes. I also prepared the manuscript with contribution from two other authors. 71 Chapter 3 Microbial ecology of the murine gut associated with the development of Dextran Sodium Sulfate-Induced colitis. Abstract and Key Words: Background: Dextran sodium sulfate (DSS) is used to induce murine colitis. Although the exact mechanism by which DSS administration causes disease is unknown, evidence suggests that resident bacteria play a role in the development of murine DSS colitis, analogous to their role in human inflammatory bowel diseases. Methods: C57BL/6 mice received 5% DSS in drinking water, and were euthanized 3 days and 14 days after initiation of DSS treatment. Cultureindependent methods were used to follow changes in community structure of the gut’s microbiota following DSS treatment. Histologic evidence of disease and changes in host gene expression were assessed. Results: Histologic colitis was minimal in DSS-treated animals at three days, but severe after 14 days. Analysis of 16S rRNA-encoding gene clone libraries demonstrated that microbial communities in the ceca of DSS-treated mice were distinct from those in control mice. The microbiota in the cecum of DSS-treated 72 animals was characterized by an overall decrease in microbial richness, an increase in members of the phylum Verrucomicrobia, and decrease in Tenericutes. Changes in the host’s inflammatory response and microbial communities occurred before histologic appearance of severe disease in the colon, but were seen concurrently in the cecum. Conclusion: DSS administration is associated with reproducible changes in the gut microbial diversity of mice. Changes in the microbial community and increases in host mediators, such as TNFα and arginase, appeared before development of severe inflammation in the cecum. This indicates that these changes in the microbial community may play role in potentiation of the abnormal inflammatory response seen in DSS-treated animals. Key words: Microbiota, DSS, 16S rRNA-encoding gene, ecology, colitis 73 Introduction: Microbes are essential to the health and well-being of their hosts (1, 2). However, presence of bacteria alone is not enough to impart these benefits, rather composition and relative abundance of specific microbes have an important role in maintaining health (3). Alterations of community structure of the indigenous microbiota have also been implicated in development of disease. One such condition in which resident bacteria are thought to play a critical role in pathogenesis is inflammatory bowel disease (IBD) (3, 4). Dysbiosis, defects in immunoregulation and defects in the barrier function are all thought to contribute to onset of disease (5-7). Evidence of a key role for the microbiota in pathogenesis is provided by studies that demonstrate that antibiotics can reduce or prevent inflammation both in patients and in murine models of disease (8, 9). In IL-10-/- mice, which develop colitis after infection with certain bacteria or in the setting of certain housing conditions, treatment with antibiotics is associated with alterations in the microbial gut community (10), and prevention or amelioration of disease (11). Of the multiple murine models of IBD, one commonly used system involves administration of dextran sodium sulfate (DSS) which induces disease very similar to human ulcerative colitis (12). Antibiotic administration has been shown to ameliorate DSS-induced colitis (13), and cathelicidin, an antimicrobial peptide, was also found to have protective effects in this model of colitis (14), indicating that the microbiota plays a role in this disease model system. 74 Multiple hypotheses have been postulated for the mechanism by which DSStriggers gut mucosal inflammation but the exact pathogenesis remains unclear (14, 15). Given that human IBD is associated with an altered microbial diversity (5, 16) and evidence for a role of the microbiota in DSS-induced colitis (14, 17), we hypothesize that this model is also associated with an altered diversity of the intestinal microbiota. To test whether DSS treatment can alter the microbial community diversity, we employed molecular techniques targeting the 16S rRNAencoding gene to follow shifts in community structure of gut bacteria in animals receiving DSS. We correlated changes in community structure with development of disease. Materials and Methods Animals: C57BL/6 mice from a breeding colony initially established with breeding stock from Jackson Laboratories (Bar Harbor, ME) were used for experiments. Animal studies were conducted at Michigan State University and were approved by the Michigan State University Animal Use Committee. Mice were housed with autoclaved bedding, given sterile food and water ad libitum, and exposed to 12:12 h light:dark cycles. Mice, between 12 to 16 wks old, were assigned to cages according to gender. Dextran sodium sulfate (DSS) (36 00050 000 MW, ICN Biochemicals Inc., CA.) was administered as a 5% solution in drinking water (8). Three groups of mice were used in this experiment: ten control mice that were maintained on sterile drinking water; ten mice that were placed on 5% DSS in 75 sterile drinking water for 3 days before being euthanized; and eleven mice that were given 5% DSS for 14 days before euthanasia (18). Necropsy and Histology: Mice were euthanized by CO2 asphyxiation and the cecal tissue harvested as described previously (19). Tissues were gently washed with 1X phosphate-buffered saline (PBS) to remove fecal contents, cut into sections and snap-frozen in liquid nitrogen. One of these sections was used for terminal restriction fragment length polymorphism (T-RFLP) analyses and clone library analysis, and another for RNA extraction. The remainder of the cecum was processed for histology as follows: Luminal contents were removed, and the tissue was washed with PBS, placed in tissue cassettes and submerged in 10% formalin for 24 hours. Tissue cassettes were transferred to a 60% ethanol solution and then processed for paraffin embedding and staining with haematoxylin and eosin (H&E). Scoring was completed using the colitis index histological scoring system used by Berndt et al. (20) and adapted from Rachmilewitz et al. (21). Briefly, sections assessed on inflammation, transmural infiltration, cell wall thickening and bleeding; and scored on a scale ranging from 0 to 40. RNA extraction and PCR array analysis: Total RNA from cecal tissue was isolated using TRIzol® (Invitrogen, Carlsbad, CA) as directed by the manufacturer’s protocol. The High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, CA) was used to convert the total RNA to 76 cDNA. Changes in host gene expression were measured using an array of genespecific primers from Superarray® (Frederick, MD), designed for the following targets (22): Jun, Cd80, Cd209a, Il12b, Irak3, Smad3, Arg1, Cd86, Mapk3, Il17a, Mapk8, Tgfb1, Cd274, Creb1, Mapk1, Il1b, Tlr2, Tlr5, Vtcn1, Cx3cr1, Foxp3, Il2, Tlr4, Tlr9, Ccr2, Cxcl1, Tnfrsf18, Il23a, H2-DMb1, Tnf, Ccr7, Cxcl10, Infg, Il4, Nfkb1, Vegfa, Itgax, Cxcl5, Il10, Il6, Mapk14, Gapdh, Cd40, Cxcl2, Il12a, Irak4, Pik3r1, ActB. The neutrophil marker, Ly-6G, was also quantified using the primer set from Sasmono et al. (23) TGGACTCTCACAGAAGCAAAG-3' and (Ly-6G forward reverse primer primer 5'5'- GCAGAGGTCTTCCTTCCAACA-3') as well as the Gapdh primer set from Cui et al. (24) (forward primer 5'-ACCACAGTCCATGCCATCAC-3' and reverse primer 5'-TCCACCACCCTGTTGCTGTA-3). Quantization was performed using LightCycler® 480 SYBR Green I Master and analyzed on a LightCycler® 480 system (Roche Diagnostics, Indianapolis, IN) as directed by manufacture’s protocol, with the following cycling conditions: 95oC activation for 10 min; 40 cycles of 95oC denaturation for 15s, and 60oC annealing for 1 min. Resulting threshold values were analyzed by calculating the 2- ΔΔ Ct values, using GAPDH as reference, to find fold regulation compared to no-DSS controls (25-27). DNA extraction: Genomic DNA was extracted from tissue using the Qiagen DNeasy® Blood & Tissue kit (Cat. No.69504, Valencia, CA). Briefly, tissue was incubated overnight in lysis buffer and proteinase K at 56 oC. The following day, this enzyme was denatured at 95 oC and DNA was purified using ethanol through 77 filter columns, as directed by the manufacturer’s protocol, and eluted in 30µl of elution buffer. This genomic DNA was then used in preparation of clone libraries and T-RFLP. Clone library construction: For clone library construction, reaction was set up as described previously (28), with illustra PuReTaqTM Ready To Go PCRTM beads (GE Healthcare, Piscataway, NJ). Briefly, amplification by polymerase chain reaction was performed using broad-ranged primers, (8F, 5'- AGAGTTTGATCCTGGCTCAG-3'; 1492R, 5'-GGTTACCTTGTTACGACTT-3'). Amplicon purification was done using a commercial kit (GFX, GE Healthcare, Piscataway, NJ) as directed by the manufacturer. Products were ligated into TOPO 4® vector (Invitrogen K4575-01, Carlsbad, CA) according to manufacturer’s specifications, and transformed into Escherichia coli. Colonies were picked into Luria Broth (LB) with carbenicillin (50 µg/ml)) and grown overnight at 37 o C. Vector specific CAGTCACGACGTTGTAAAACGACGGC-3'; primers and (M13F, M13R, 5'5'- CAGGAAACAGCTATGACCATG-3') were used to screen these colonies for bacterial clones containing appropriate 1.5 KB amplicon insert. Partial 16S sequences were determined by a single sequencing run using the 8F primer, at the Genomic Core at Michigan State University. Raw sequence data were processes through an automated “information pipeline” available through the Ribosomal Database Project (RDP) Web site (http://rdp.cme.msu.edu/). Data were screened using the Chimera Check Program before uploading into the 78 RDP. Following alignment of sequences via myRDP (29), distance matrices representing each of the libraries were downloaded and then taxonomic assignments designated (80% confidence cut-off) using Classifier through the RDP website. These distance matrices were also input into mothur (30) to be grouped into operational taxonomic units (OTUs). Analyses were performed using a 97% sequence similarity to denote species level. An input table was also generated for the EstimateS program via the RDP Pipeline. EstimateS (31) was used to calculate ecological diversity indices from aligned sequences. Dendrograms based on the Bray-Curtis similarity index were constructed using the Mega3 program (32). 16S small subunit rRNA gene sequences obtained by clone library analysis were subjected to in silico terminal restriction fragment length polymorphism (T-RFLP) analysis using the TRF-cut program in the ARB suite of programs (33). The MspI enzyme, a four base cutter, was used to calculate predicted terminal restriction fragments (TRFs) from clone sequences. Histograms were then constructed displaying relative abundances of these in silco generated TRFs and compared to actual traces retrieved from T-RFLP analysis described below. Fragment sizes obtained by T-RFLP anlaysis were compared to sizes of the TRFs generated in silico from cloned 16S sequences. These fragments were identified within two basepairs (± 2 bp) of predicted TRFs (34). T-RFLP analysis: T-RFLP was performed as described elsewhere (35) using primers 1492R and FAM labeled 8F. In summary, genomic DNA was amplified 79 using the aforementioned primers with PCR conditions: 94 oC denaturation for 2 min; 30 cycles of denaturation at 94 oC for 30 s, annealing at 58 oC for 45 s, and extension at 72 oC for 90 s; and final extension at 72 oC for 4 min. The 1.5 kb PCR product was verified and purified with GFX columns (GFX, GE Healthcare, Piscataway, NJ), then subjected to digestion with MspI restriction enzyme for two hours. Digested DNA was submitted for analysis to the Genome Technology Support Facility (GTSF) at Michigan State University. Traces were visualized using the program Genescan (Applied Biosystems, Foster City, CA). The T-RFLP Stats collection of programs (http://styx.ibest.uidaho.edu/ibest/research.html) (36) were used to analyze traces from T-RFLP output data. Briefly, peaks of each trace were binned into operational taxonomic units (OTU) to produce data categorized by both OTUs and abundance. This was then used as input for the EstimateS (http://viceroyeebuconnedu/estimates) (31) program where the Bray-Curtis values were calculated and used to compare the diversity among communities. The Mega3 program (32) was used to construct dendrograms showing the relationship between community structures using the Bray-Curtis values. Significance differences between community structures were calculated using the parsimony test from the mothur suite of programs. qPCR analysis of microbial communities: The quantity of 16S rRNA operons in samples relative to a single-copy host gene was measured using a 80 primer/probe set that targets a broad range of rRNA-encoding gene sequences (5’-TCCTACGGGAGGCAGCAGT-3’), reverse GGACTACCAGGGTATCTAATCCTGTT-3’), and primer probe (5’- (5’-[6-FAM]- 203CGTATTACCGCGGCTGCTGGCAC-[TAMRA]-3’) (37). A primer/probe set targeting a 264-bp portion of the TNF alpha gene was used as a reference using 200 nanomoles of forward (TNFα_mu_se; 5’-GGCTTTCCGAATTCACTGGAG3’) and reverse primers (TNFα_mu_as; 5’- CCCCGGCCTTCCAAATAAA-3’), and 100 nanomoles of probe (TNFα_mu_probe; 5’-[Cy5]- ATGTCCATTCCTGAGTTCTGCAAAGGGA-[Iowa Black RQ™]-3’) 213 (38). The reaction mix consists of LightCycler® 480 Probes Master reaction mix (Roche) at 1X concentration, and appropriate primer/probe pair. Amplification of each gene was done under separate run conditions: cycling conditions for the 16S target involved an activation step of 50 oC for 2 min followed by 95 oC for 10 min. Fortyfive cycles were done at 95 oC for 15 s and 60 oC for 1 min before holding. For the TNF reference gene cycling conditions included an activation step of 50 oC for 2 min followed by 95 oC for 10 min and forty-fives cycles of 95 oC for 20 s and 64 oC for 30 s. Calculations of 2-ΔΔCt were made to compare changes in the amount of 16S from samples between treatment groups (21,22). Statistical Analysis: The non-parametric Kruskal-Wallis test was used to analyze histological scores. Differences in abundances of specific operational taxonomic units among treatment groups were analyzed by ANOVA. Statistical differences in dendrograms comparing distances between microbial communities 81 were calculated using the parsimony test function in mothur (30). Probability values less than 0.05 were considered significantly different. Results DSS treatment leads to inflammation of the murine intestinal tract. Wildtype mice were treated with 5% DSS in drinking water and gastrointestinal tissue examined histologically after 3 and 14 days of treatment. All animals survived DSS treatment and no significant clinical signs were noted. A mild inflammatory infiltrate developed in the cecum 3 days after DSS administration (figure 3.1 b), with notable progression of disease after 14 days characterized development of a massive inflammatory cell infiltrate and severe edema (figure 3.1 c). These changes were significantly different from the control state (p<0.05), as well as between 3 day and 14 day treatment outcomes (figure 3.1 g). Similarly in the colon, a few animals developed morphological and inflammatory changes after 3 days of DSS treatment, but as a group, these changes were not significant when compared to controls. However, following 14 days of DSS administration colonic tissue showed evidence of severe inflammation and edema which was significantly greater than controls of animals after 3 days of treatment (figure 3.1 h). 82 50 µm b c d e f * 40 30 20 10 0 * 40 ns ** * 20 Cecum h da y D SS 14 3 da y no D SS D SS da y 14 3 da y D SS 0 D SS 10 N o g ** 30 D SS Inflammation a Colon Figure 3.1. Histopathology in DSS-treated mice. Hemotoxylin and eosin (H&E) stained sections were prepared from cecum sections of (a) untreated control mice (b) mice after 3 days of DSS treatment and (c) after 14 days of DSS. Arrow indicates submucosl edema. H&E sections were also prepared from colon samples of (d) untreated controls (e) mice after 3 days of DSS treatment (arrow points to inflammation) and (f) animals after 14 days of DSS (arrow shows abscess). Histopathologic scores were calculated for sections from all 31 animals for (g) cecum and (h) colon sections. Statistical analysis done was Kruskal-Wallis test. *p< 0.05 **p<0.001. Initial magnification 40X. 83 Changes in host gene expression following DSS treatment Using a quantitative PCR array, we found significant changes in expression of ten host genes in cecal tissue following DSS treatment (figure 3.2, supplemental figure 1). Expression of arginase 1 began to increase after 3 days and was significantly up-regulated ~seven fold (p<0.05) after 14 days of DSS administration. A significant increase in tumor necrosis factor-alpha (TNFα) was also seen after 3 days of DSS treatment, with a 12.9 fold up-regulation (p<0.05), and remained significantly up-regulated at the 14-day time point. Eight other genes were down-regulated, compared to untreated controls. They were TLR 5, IL17, Ccr2, Cx3cr1, Cxcl2, Cd40, Cd80 and Cd209a. TLR 5 had significant down-regulation after 14-days of DSS exposure, as did Cd40 and Cxcl2. Cd80 showed significant change after 3 days of DSS only marginal change after 14 days. Ccr2, Cx3cr1, Cd209a and IL17a showed significant downregulation both at the 3 day and 14 day time points. Presence of neutrophils, as indicated by measuring the Ly-6G marker, increased significantly with DSS treatment with a 7.7±1.2 fold increase after 3 days of DSS administration, and 14.0±1.8 fold increase after 14 days of DSS (supplemental table 3.1). 84 Fold regulation compared to No-DSS Control 15 * * 10 * 3 Day DSS 14 Day DSS 5 0 -5 -10 TLR5 Ccr2 IL17a Cd80 Cd40 Cd209 TNF Cx3cr1 Cxcl2 Arg1 * * * * * * * * * * * * Figure 3.2. Changes in host gene expression after 3-days (dark bars) or 14-days (grey bars) ± standard deviation of DSS administration. Expression levels were compared to expression in tissue from animals not exposed to DSS, after normalization with GAPDH. Statistical analysis done was Student’s ttest. Statistically significant differences (p<0.05) are denoted by asterisks (*). 85 Shifts in gut microbial diversity following DSS treatment. To obtain an overview of the status of the microbial community present in the gastro-intestinal tract of each of the animals in this study, 16S terminal restriction fragment length polymorphism (T-RFLP) analysis was performed on DNA extracted from cecal tissue. Analysis of microbial communities in the cecum of all 31 animals in the study revealed that the structure of the microbial communities from DSS treated mice were significantly different from the no-DSS controls (p<0.001) (supplemental figures 3.2). Since T-RFLP provides a broad overview of community structure without information regarding changes in specific organisms, 16S clone libraries were constructed to further investigate the microbial community in four representative mice from each treatment group. A total of 328 partial 16S rRNA-encoding gene sequences were retrieved from control animals (71, 84, 89 and 84 per animal), 364 sequences from animals 3 days after initiation of DSS treatment (88, 92, 93 and 91) and 370 from animals treated with 14 days of DSS (92, 94, 89 and 95). Clone library analysis confirmed T-RFLP findings, indicating that community compositions of the microbiota in DSS treated animals were more similar to each other than they were to those of control mice (figure 3.3, supplemental figure 3.4). T-RFLP traces were compared to in silico TRFs of clone libraries to verify that OTUs matched the traces (supplemental figures 3.5- 3.8). Communities in all DSS-treated animals were significantly different from those in no-DSS controls (p<0.001 by the parsimony test implemented in the analysis suite mothur (30)). Rarefaction analysis of clone libraries revealed that DSS treatment resulted in a 86 decrease in phylotype richness (supplemental 3.4). This finding was also supported by a decrease in the Simpson diversity index (1/D) following DSS treatment from 43.0±12.1 to 21.3±8.3 (3 days) and 19.7±7.0 (14 days) (p<0.05). However, quantitative PCR of the 16S gene from each treatment group showed that there were no significant changes in overall bacterial biomass relative to noDSS controls (fold change for 3 day DSS= 0.68±0.92 SD, 14 day DSS= 0.97±0.82 SD). 87 * 0.40 0.30 14 Day DSS 0.20 3 Day DSS 0.10 0.00 No DSS Figure 3.3. Comparison of cecal communities in control animals (black triangles) and in animals following 3 days of DSS treatment (white squares) and 14 days of treatment (black squares). Using an OTU definition of 97% similarity, the Bray-Curtis similarity metric was calculated for each pair-wise comparison and then results displayed in denrogram format. Analysis by parsimony tests indicate that there is a statistically significant (*p<0.001) difference between the communities in control animals and in both groups of DSS-treated animals. 88 We analyzed changes in the relative abundance of specific phylotypes by classification of the 16S rRNA-encoding gene sequences. DSS treatment resulted in a decrease in members of the phyla Bacteriodetes and Tenericutes. Alterations in the microbial community were also apparent at lower taxonomic levels. For example, there was an increase in unclassified genera under the family Lachnospiraceae (figure 3.4, supplemental 3.3). The majority of classified phylotypes in animals from all three experimental groups were members of the phylum Firmicutes, which include the classes Clostridia and Bacilli. Bacteriodetes was most abundant in control animals and represented to a lesser extent in DSS treated groups (figure 3.4 A, supplemental 3.3). A striking observation was that members of the phylum Verrucomicrobiae were detected only in DSS treated mice and not in control mice (figure 3.4 A). Among members of the phylum Bacteriodetes richness of the population did not change, indicating that changes were mainly due to shift in abundances of different bacteria within this phylum. Among the Firmicutes, unclassified genera among families Ruminoccocaceae and Lachnospiraceae were most prevalent. These groups showed treatment related shifts as seen in pie charts of figure 3.4 B. When analysis was done using 97% sequence similarity, there was high richness among Lachnospiraceae, compared to other major groups within the Firmicutes (table 3.1), indicating increased diversity within this category (p<0.05). These groups also showed treatment related shifts (table 3.1). 89 Proportion of bacteria in each treatment group A 98 96 94 92 90 88 86 84 82 80 Phylum Level Assignment Tenericutes* Crenarceota Verrucomicrobiae* Proteobacteria Bacteriodetes Firmicutes 0 No DSS Control 3 Day DSS 14 Day DSS mean proportion of bacteria per treatment group 3.14 % ±1.82 0.30 % ±0.60 0.00 % ±0.00 0.55 % ±0.67 9.39 % ±5.00 86.90 % ±7.02 0.00 % 0.00 % 2.75 % 0.28 % 0.83 % 96.15 % ±0.00 ±0.00 ±1.10 ±0.56 ±0.55 ±1.49 0.27 % 0.00 % 4.64 % 0.00 % 5.04 % 90.05 % ±0.54 ±0.00 ±3.42 ±0.00 ±4.17 ±3.31 Figure 3.4 (A). Changes in the cecal gut microbial community following DSS administration at the phylum level. Analysis was done by ANOVA and statistical significance (p<0.05) is denoted by asterisks (*). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. 90 B No DSS Control Assignments within Firmicutes 3 Day DSS 14 Day DSS mean proportion of bacteria per treatment group Lachnospiraceae (N/A) * 32.93 % ±9.22 Ruminococcaceae (N/A) 26.43 % ±9.58 Clostridiales (N/A) 10.30 % ±7.49 Lachnospiraceae Incertae Sedis 10.10 % ±1.53 59.63 % ±6.27 16.43 % ±7.13 8.20 % ±5.62 8.32 % ±7.17 57.69 % ±11.64 17.39 % ± 5.74 8.95 % ± 4.03 8.75 % ± 3.46 * denotes statistical significance (p<0.05) Figure 3.4. (B) Changes in the cecal gut microbial community following DSS administration within the phylum Firmicutes. Analysis was done by ANOVA and statistical significance (p<0.05) is denoted by asterisks (*). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. 91 Shared OTUs among all groups OTUs in control group only OTUs in 3 day DSS group only OTUs in 14 day DSS group only Shared OTU in DSS treated groups Clostridiales N=17 (%) 6 (35) 4 (24) 2 (12) 2 (12) 3 (18) Ruminococcus N=36 (%) 16 (44) 8 (22) 4 (11) 5 (14) 3 (8) Lachnospiraceae N=65 (%) 15 (23) 13 (20) 8 (12) 12 (18) 17 (26) Table 3.1 Showing distribution of OTU’s across treatments from major groups within the Firmicutes. 92 Discussion: Indigenous gut microbiota are felt to play a key role in pathogenesis of inflammatory bowel disease. Much of the evidence for the involvement of intestinal bacteria in IBD comes from studies with murine models of disease (39, 40). Many of these models have specific alterations in host defenses ranging from altered epithelial cell function to altered innate or adaptive immunity (41-44). These alterations in host defense result in abnormal interactions between the host and indigenous microbiota that lead to disease (45). Evidence for this comes from the fact that antibiotic administration reduces severity of disease in these models and rederivation of these mice to the germ-free state prevents initiation of disease (13, 46). Administration of DSS consistently triggers intestinal inflammation in rodents. Similar to other models, antibiotic administration has a beneficial effect in the system again pointing to a role for the indigenous microbiota in disease development (47). Effects of DSS on germfree mice have been variable. In some strains of mice more severe colitis is encountered while early death, without development of significant inflammation, is observed in others (48, 49). Germfree IQI/Jic mice have a high mortality rate, compared to conventional mice, when given a concentration of 5% DSS in drinking water (45). These mice died secondary to hemorrhage within three days after DSS treatment, indicating a protective function of the microbiota against toxic effects of DSS on the gut epithelium. These results are consistent with the finding that antibiotic 93 administration or genetic defects in TLR signaling also increased mortality in DSS-treated conventional mice (50, 51). Differences in these two studies employing germ-free mice likely reflects differential effects of two doses of DSS on the epithelium, but may also reflects differences in composition of the microbiota of conventional control animals. Since specific subsets of the “normal” microbiota have differing ability to initiate or sustain experimental colitis (47, 52) these results emphasize the importance of characterizing changes in the gut community associated with DSS-induced colitis across time. Although it was originally suggested that IBD resulted from infection with an unknown bacterial pathogen (53), the more contemporary view is that entire shifts in the microbial community structure may be the trigger for disease (5). An altered gut microbial community has been associated with disease as shown in human studies and animal models (54, 55), suggesting that there is a change in abundance of bacteria that are necessary for maintaining homeostasis (54, 55). After administering DSS to mice, we show that treatment results in changes in microbial community structure in the gut, also noted by others (51). Additionally, we found that these changes occur early (within 3 days) and are characterized by reduced overall diversity while bacterial load remains unchanged. Some community members are below limits of detection after DSS treatment, while others increase in abundance demonstrating a selective effect of DSS treatment on specific members of the community. It has been shown that certain bacteria can de-polymerization DSS and thus can grow in a DSS-rich environment better than others (56). This may explain the bloom of Verrucomicrobia that we 94 observed in DSS treated mice. This group of microbes, which has been found in mammalian intestine at very low abundances (57), metabolize sulfur and degrade mucin (58). Therefore, perturbations of the community can cause previously underrepresented members of the community to become dominant (59), resulting in shifts in community structure, as seen in the phylum Bacteriodetes and families Ruminococcaceae and Lachnospiraceae. Another study has profiled effects of DSS treatment on the microbial community (51). Similar to our study, it was noted that shifts in relative abundances of specific phylotypes were observed following DSS treatment, although specific changes were different from what we observed. One obvious reason for these differences could be explained by the fact that different anatomic locations were sampled (cecal mucosa used here versus distal colonic contents in study by Heimesaat et al. (51)), as well as length and concentrations of DSS administration. Additionally, it is likely that the baseline microbial communities were quite different between the animals in that study and the animals we used. Recent reports have demonstrated that the baseline microbiota can differ in genetically identical animals obtained from different sources and these differences can have a profound effect on host immune responses (60, 61). In our study, disease severity in the cecum increased after 3 days and 14 days post DSS administration, concurrent with changes observed in the microbial community, indicating a link between the changes in diversity of the microbiota and disease. Since shifts in the community structure can reduce beneficial members of the indigenous microbiota that act to maintain epithelial health, such 95 as production of short-chain fatty acids and stimulation of mucin production (55, 62-65), it is likely that such changes can affect host inflammatory responses (45, 66). Others have suggested that DSS-induced colitis occurs due to breaches in the epithelial barrier with exposure to antigens produced by luminal microbes (5, 6773). However, there is evidence that detection of an altered microbial community is an important factor in host response (74, 75). Our data indicate that DSS induces changes in the microbiota, and together with a compromised barrier, appears to facilitate disease initiation and progression. It has been shown that the host responds to microbial-associated molecular patterns (MAMPS) via tolllike receptors (TLRs) in development of colitis (17, 40, 76). We found significant a change in expression of TLR 5, expression of which was decreased as colitis progressed. It has been previously demonstrated that TLR 5 is down-regulated in patients with severe ulcerative colitis (77), in concurrence with our data. However, the role of flagella in DSS colitis is not dependent on TLR5 (40, 78), suggesting that down-regulation is perhaps a response from over-stimulation from microbes through a deteriorating epithelial layer. It is noteworthy that we saw an increase in arginase 1 after 14 days of DSS treatment. This could be due to an increase of neutrophils after DSS administration, as indicated by increased expression of the neutrophil marker Ly6-G. Arginase 1 also has a protective role in the Citrobacter rodentium model of colitis where this enzyme inhibition aggravated disease (79). It has been seen that arginase expression increases with TNFα/lipopolysaccharides (LPS) 96 stimulation in human intestinal microvascular endothelial cells (80). With breaches in the epithelial layer that allow increased stimulation by LPS, together with an increase in TNFα as seen here and in other studies with DSS (70, 81), it follows that arginase expression is likely to be up-regulated. Interestingly, although TNFα appeared to be at lower levels at 14 days compared to 3 days, arginase was up-regulated at 14 days compared to 3 days, consistent with the presumed role for this gene product in the epithelial repair response. This is not unexpected since DSS administration disrupts the epithelial barrier (15). To our knowledge, this study is the first to document the significant increase of this enzyme in DSS-induced colitis. Although others have noted changes in the gut microbiota following DSS treatment (51) after development of disease, this is the first study to monitor DSS-induced changes in the intestinal microbiota over time. In this study, we not only corroborate that changes in the gut microbial community are associated with DSS-induced disease, but we also show that diversity of the microbiota changed as early as after 3 days and continued to 14 days of DSS administration. Early shifts in the microbial community were characterized with increases in abundances of microbes previously undetectable by methods used. These altered communities are present at onset of inflammation, and maintained as inflammation severity increased, reinforcing the idea that disease occurs, and persists, in the presence of an altered intestinal community. These early changes in the microbiota support the view that disturbance of the resident bacterial 97 community structure may have a very important role in onset of diseases, such as IBD. Acknowledgment I would like to extend thanks to Jason Pratt for technical assistance, and Dr. Courtney Robinson, Dr. Grace Chen and Dr. Gary Huffnagle for helpful discussions and review of the manuscript. I am especially grateful for the funding provided by the Center for Microbial Pathogenesis at MSU and the NIH. I would also like to acknowledge the Genomics Core at Michigan State University where all the sequencing and T-RFLP data were generated, as well as the RDP staff for facilitating the work. 98 SUPPLEMENTAL INFORMATION Table 3.2. Changes in host gene expression following DSS treatment. TARGET FUNCTION FOLDREGULATION 3-Day 14-Day DSS DSS Th1 response Interleukin 12a (Il12a) differentiation of naïve T cells Interleukin 12b (Il12b) differentiation of naïve T cells Interleukin 2 (Il2) leukocytotrophic hormone Tumor Necrosis Factor alpha (Tnf) Interferon gamma (Infg) proinflammatory cytokine Toll-like Receptor 2 (Tlr2) pathogen recogition Toll-like Receptor 4 (Tlr4) pathogen recognition Toll-like Receptor 5 (Tlr5) pathogen recognition Toll-like Receptor 9 (Tlr9) pathogen recognition Chemokine (C-X-C motif) ligand 10 (Cxcl10) chemoattractant 2.35 ± 0.81 1.42 ± 0.33 1.79 ± 0.00 12.91* ± 1.23 2.26 ± 0.20 -1.22 ± 0.91 1.21 ± 1.28 -2.38 ± 1.15 1.05 ± 1.41 2.76 ± 2.90 1.57 ± 1.66 -2.53 ± 0.40 -2.45 ± 0.10 10.51* ± 1.62 -2.05 ± 0.12 -2.51 ± 0.38 1.74 ± 1.31 -3.60* ± 1.51 -1.58 ± 1.53 2.64 ± 1.31 1.82 ± 0.00 -8.19* ± 1.29 -2.26 ± 0.26 -4.33* ± 1.28 -3.60* ± 0.58 -9.00* ± 0.89 2.48 ± 0.80 -2.02 ± 0.13 Th2 response Interleukin 4 (Il4) chemokine (C-C motif) receptor 2 (Ccr2) Th17 response Interleukin 17a (Il17a) Interleukin 23a (Il23a) proinflammatory cytokine differentiation of naïve T cells chemoattractant induction and mediation of proinflammatory responses (allergic) differentiation of naïve T cells 99 Table 3.2 (cont’d) CD4+ T cell marker Interleukin 6 (Il6) Mitogen-activated protein kinase 8 (Mapk8) Cluster of Differentiation 80 (Cd80) Cluster of Differentiation 86 (Cd86) chemokine (C-C motif) receptor 7 (Ccr7) V-set domain-containing T-cell activation inhibitor 1 (Vtcn1) Tumor necrosis factor receptor superfamily member 18 (Tnfrsf18) "Integrin, alpha X (Itgax/CD11c)" Tregs Transforming growth factor beta (Tgfb1) Interleukin 10 (Il10) forkhead box P3 (Foxp3) Transcription factors nuclear factor kappa-lightchain-enhancer of activated B cells (Nfkb1) Jun pro-inflammatory and antiinflammatory "regulation of cellular activities (gene expression, mitosis, differentiation, proliferation, cell survival/apoptosis)" receptor -1.41 ± 0.26 -1.83 ± 0.35 -2.94 ± 0.97 -1.75 ± 1.03 -3.59* ± 0.13 1.41 ± 2.10 -2.71 ± 0.43 1.61 ± 0.00 -2.45 ± 1.38 2.23 ± 1.73 -2.42 ± 1.97 -2.8 ± 0.00 receptor 1.21 ± 0.66 1.42 ± 1.69 receptor 1.47 ± 0.44 1.26 ± 2.05 differentiation of naïve T cells/anti-inflammatory cytokine anti-inflammatory cytokine differentiation of naïve T cells/anti-inflammatory cytokine -1.4 ± 0.53 -2.85 ± 0.00 -1.6 ± 1.24 1.01 ± 1.24 -2.78 ± 1.06 -2.68 ± 1.02 regulation of the immune response to infection -1.19 ± 0.56 -1.1 ± 1.92 early response factor to AP-1 1.43 ± 1.47 -1.33 ± 0.90 -1.24 ± 0.90 1.62 ± 0.91 -1.1 ± 1.91 -1.17 ± 1.46 1.65 ± 1.84 1.94 ± 1.36 receptor receptor ligand on antigen presenting cells Smad3 Interleukin-1 receptorassociated kinase 3 (Irak3) Interleukin-1 receptorassociated kinase 4 (Irak4) signalling innate response signalling innate response 100 Mitogen-activated protein kinase 14 (Mapk14) "regulation of cellular activities (gene expression, mitosis, differentiation, proliferation, cell survival/apoptosis)" Mitogen-activated protein "regulation of cellular activities kinase 1 (Mapk1) (gene expression, mitosis, differentiation, proliferation, cell survival/apoptosis)" Mitogen-activated protein "regulation of cellular activities kinase 3 (Mapk3) (gene expression, mitosis, differentiation, proliferation, cell survival/apoptosis)" CAMP responsive element binds DNA to regulate binding protein 1 (Creb1) transcription B cell Phosphatidylinositol 3kinase regulatory subunit alpha (Pik3r1) Cluster of differentiation 40 (Cd40) Other Cluster of differentiation 209a (Cd209a) Arginase 1 (Arg1) Table 3.2 (cont’d) -1.29 ± -1.35 ± 0.38 1.84 -1.27 ± 0.31 -1.03 ± 2.32 -1.49 ± 1.61 -1.57 ± 1.85 -1.45 ± 0.25 -1.32 ± 1.76 insulin receptor -1.38 ± 0.64 -1.14 ± 1.16 receptor -2.87 ± 1.28 -4.98* ± 1.31 receptor -3.03* ± 0.30 2.59 ± 0.45 2.58 ± 1.13 -6.31* ± 1.30 2.23 ± 0.00 2.23 ± 1.60 -2.4 ± 0.18 1.71 ± 0.20 -1.08 ± 1.49 7.7*± 1.20 -5.86* ± 1.05 7.22* ± 1.46 1.23 ± 1.11 -3.00* ± 1.79 -1.93 ± 0.13 2.94 ± 2.13 -2.65 ± 1.93 -2.42 ± 0.29 -3.46* ± 0.84 14.0*± 1.80 wound healing Cluster of differentiation 274 (Cd274) Cx3cr1 receptor Chemokine (C-X-C motif) ligand 1 (Cxcl1) MHC class II beta chain (H2-DMb1) Vascular endothelial growth factor A (Vegfa) Chemokine (C-X-C motif) ligand 5 (Cxcl5) Chemokine (C-X-C motif) ligand 2 (Cxcl2) Lymphocyte antigen 6 complex (Ly-6G) chemoattractant receptor antigen processing and presentation cell differentiation, maturation, proliferation, anti-apoptosis chemoattractant chemoattractant Neutrophil marker 101 Table 3.3. Taxonomic distribution of rRNA genes clones from RDP classifier analysis of treatment group 3 14 Day Day No PHYLUM CLASS FAMILY GENUS DSS DSS DSS Total Bacteroidetes 33 3 19 Bacteroid etes Bacteroid etes Porphyrom onadaceae Tannerella 13 2 10 Parabacteroides 1 2 17 0 1 0 0 2 7 1 0 0 282 351 334 7 2 4 0 4 1 4 9 0 78 1 6 0 1 58 0 4 0 0 58 0 30 2 0 1 0 95 29 0 1 0 6 209 29 0 0 0 6 193 1 2 0 1 0 4 2 26 3 0 30 1 0 30 0 N/A Crenarcha eota N/A Total Firmicute s Firmicutes Clostridia Ruminococ caceae Anaerotruncus Papillibacter Ruminococcace ae Incertae Sedis Ruminococcus Fastidiosipila N/A Sporobacter Lachnospir Lachnospiracea aceae e Incertae Sedis Roseburia Lachnospira Catonella Butyrivibrio N/A Peptococc aceae Peptococcus N/A Peptostreptococ Peptostrep caceae Incertae tococca Sedis N/A 102 Table 3.3 (cont’d) Bacilli Lactobacill aceae N/A Erysipelotr Erysipelotri chaceae ichi Lactobacillus 2 2 1 0 0 0 Allobaculum Turicibacter Erysipelotrichac eae Incertae Sedis Holdemania Erysipelotrichac eae Incertae Sedis 12 4 2 0 0 3 0 0 1 1 1 0 0 6 2 0 0 1 1 0 0 Herbaspirillium 1 1 0 0 0 1 0 0 0 Anaeroplasma 11 0 1 Akkermansia 0 1 328 10 0 365 17 0 371 N/A Total Proteobacteria Proteobac teria Betaprote obacteria Oxalobacte raceae N/A N/A Tenericut es Mollicute s Anaeropla smatacea e Verrucom Verrucom Verrcomic icrobiae icrobia robiaceae N/A TOTAL 103 * ** 0.25 0.20 14 Day DSS 0.15 0.10 0.05 3 Day DSS 0.00 No DSS Figure 3.5. Comparison of the cecal community in control animals (black triangles), and animals following 3 days of DSS treatment (white squares) and 14 days of treatment (black squares). Using T-RFLP, the Bray-Curtis similarity metric was calculated for each pair-wise comparison and then the results displayed in dendrogram format. Communities that were further analyzed, by construction of clone libraries, are indicated by arrows. Analysis by the parsimony test indicates that there is a statistically significant difference (*p<0.05 and **p<0.001). 104 140 120 OTU 100 80 60 40 14 day 3 day Control 20 0 0 50 100 150 200 250 300 350 400 Collection effort/ Sequences Figure 3.6. The rarefaction curves at 97% sequence similarity comparing the microbial communities in mice after 3-days of DSS treatment (pink), 14-days of DSS treatment (blue) and the controls (yellow). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. 105 in silico TRFs Relative flourescence units T-RFLP traces of control communities size (base pair) Figure 3.7. Comparisons between T-RFLP traces and histograms of the terminal restriction fragments (TRFs) of in silico digest of the clone library constructed from the control communities. 106 Relative flourescence units T-RFLP traces of 3 day DSS communities in silico TRFs size (base pair) Figure 3.8. Comparisons between T-RFLP traces and histograms of the terminal restriction fragments (TRFs) of in silico digest of the clone library constructed from the 3-day DSS administered communities. 107 Relative flourescence units T-RFLP traces of 14 day DSS communities in silico TRFs size (base pair) Figure 3.9. Comparisons between T-RFLP traces and histograms of the terminal restriction fragments (TRFs) of in silico digest of the clone library constructed from the 14-day DSS administered communities. 108 TREATMENT PHYLUM GENUS Control Tannerella (1) Coprococcus (1) Roseburia Syntrophococcus Coprococcus (2) Coprococcus (3) Oscillibacter (1) Oscillibacter (2) Tannerella (2) Coprococcus (3) Coprococcus (6) Dorea Peptococcus Akkermansia 3 Day DSS 14 Day DSS Bacteriodetes Firmicutes Firmicutes Firmicutes Firmicutes Firmicutes Firmicutes Firmicutes Bacteriodetes Firmicutes Firmicutes Firmicutes Firmicutes Verrucomicrobia TRF from clone (size/ base pairs) 83 261 76 115 197 236 259 252 58 176 142 175 258 223 TRF from T-RFLP trace (size/ base pairs) 83 260 78 116 197 235 260 253 56 174 142 174 258 223 Table 3.4. Comparison of common terminal restriction fragment lengths generated by in silico digests of clones and the terminal restriction fragment lengths from T-RFLP via Msp1 digests. 109 REFERENCES 110 REFERENCES 1. Dethlefsen L, McFall-Ngai, M. & Relman, D. A. An ecological and evolutionary perspective on human–microbe mutualism and disease. Nature. 2007;449:811-818 2. Ley RE, Peterson DA, Gordon JI. Ecological and Evolutionary Forces Shaping Microbial Diversity in the Human Intestine. Cell. 2006;24:837-848 3. Round J, Mazmanian S. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol. 2009;9:313-323 4. Subramanian S, Campbell BJ, Rhodes JM. Bacteria in the pathogenesis of inflammatory bowel disease. Curr Opin Infect Dis. 2006:475-484 5. Packey C, Sartor R. Interplay of commensal and pathogenic bacteria, genetic mutations, and immunoregulatory defects in the pathogenesis of inflammatory bowel diseases. J Intern Med. 2008;263:597-606 6. Swidsinski A, Ladhoff A, Pernthaler A, et al. Mucosal flora in inflammatory bowel disease. Gastroenterol. 2002;122:44-54 7. Ogura Y, Bonen DK, Inohara N, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature. 2001;411:603-606 8. Kang SS, Bloom SM, Norian LA, et al. An antibiotic-responsive mouse model of fulminant ulcerative colitis. PLoS Med. 2008;5:e41 9. Swidsinski A, Weber J, Loening-Baucke V, et al. Spatial Organization and Composition of the Mucosal Flora in Patients with Inflammatory Bowel Disease. J Clin Microbiol. 2005;43:3380-3389 10. Sellon RK, Tonkonogy S, Schultz M, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66:5224-5231 11. Madsen KL, Doyle JS, Tavernini MM, et al. Antibiotic therapy attenuates colitis in interleukin 10 gene-deficient mice. Gastroenterol. 2000;118:1094-1105 111 12. Okayasu I, Hatakeyama S, Yamada M, et al. A Novel Method in the Induction of reliable Experimental Acute and Chronic Ulcerative Colitis in Mice. Gastroenterol 1990;98:694-702 13. Rath H, Schultz M, Freitag R, et al. Different Subsets of Enteric Bacteria Induce and Perpetuate Experimental Colitis in Rats and MIce. Infect Immun. 2001;69:2277-2285 14. Tai EK, Wu WK, Wong HP, et al. A New Role for Cathelicidin in Ulcerative Colitis in Mice. Exp Biol Med (Maywood). 2007;232:799-808 15. Kitajima S, Takuma S, Morimoto M. Changes in Colonic Mucosal Permeability in Mouse Colitis Induced with Dextran Sulfate Sodium. Exp Anim. 1999;48:137-143 16. Frank DN, St. Amand AL, Feldman RA, et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. PNAS. 2007;104:13780-13785 17. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, et al. Recognition of Commensal Microflora by Toll-Like Receptors Is Required for Intestinal Homeostasis. Cell. 2004;118:229-241 18. Vowinkel T, Kalogeris T, Mori M, et al. Impact of Dextran Sulfate Sodium Load on the Severity of Inflammation in Experimental Colitis. Dig Dis Sci. 2004;49:556-564 19. Young VB, Knox KA, Pratt JS, et al. In Vitro and In Vivo Characterization of Helicobacter hepaticus Cytolethal Distending Toxin Mutants. Infect Immun. 2004;72:2521-2527 20. Berndt BE, Zhang M, Chen G-H, et al. The Role of Dendritic Cells in the Development of Acute Dextran Sulfate Sodium Colitis. J Immunol. 2007;179:6255-6262 21. Rachmilewitz D, Karmeli F, Takabayashi K, et al. Immunostimulatory DNA ameliorates experimental and spontaneous murine colitis. Gastroenterol. 2002;122:1428-1441 112 22. Lees CW, Zacharias WJ, Tremelling M, et al. Analysis of Germline GLI1 Variation Implicates Hedgehog Signalling in the Regulation of Intestinal Inflammatory Pathways. PLoS Med. 2008;5:1761-1775 23. Sasmono RT, Ehrnsperger A, Cronau SL, et al. Mouse neutrophilic granulocytes express mRNA encoding the macrophage colony-stimulating factor receptor (CSF-1R) as well as many other macrophage-specific transcripts and can transdifferentiate into macrophages in vitro in response to CSF-1. J Leukoc Biol. 2007;82:111-123 24. Cui W, Cuartas E, Ke J, et al. CD200 and its receptor, CD200R, modulate bone mass via the differentiation of osteoclasts. PNAS. 2007;104:14436-14441 25. Pfaffl MW. A new mathematical model for relative quantification in realtime RT-PCR. Nucleic Acids Res. 2001;29:e45 26. Guide to Performing Relative Quantitation of Gene Expression Using Real-Time Quantitative PCR. Applied Biosystems. 2004 27. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3:1101-1108 28. Young VB, Schimdt TM. Antibiotic-Associated Diarrhea Accompanied by Large-Scale Alterations in the Composition of the Fecal Microbiota. J Clin Microbiol. 2004;42:1203-1206 29. Cole JR, Chai B, Farris RJ, et al. The ribosomal database project (RDP-II): introducing myRDP space and quality controlled public data. Nucleic Acids Res. 2007;35:D169-172 30. Schloss P, Westcott S, Ryabin T, et al. Introducing mothur: Open Source, Platform-independent, Community-supported Software for Describing and Comparing Microbial Communities. Appl Environ Microbiol. 2009;75:7537-7541 31. Colwell RK. EstimateS: Statistical estimation of species richness and shared species from samples. User's Guide and application published at: http://purloclcorg/estimates. 2006 113 32. Kumar S, Tamura K, Nei M. MEGA3: integrated software for molecular evolutionary genetics analysis and sequence alignment. Brief Bioinform. 2004;5:150-163 33. Ludwig W, Strunk O, Westram R, et al. ARB: a software environment for sequence data. Nucleic Acid Res. 2004;32:1363-1371 34. Liu WT, Marsh TL, Cheng H, et al. Characterization of microbial diversity by determining terminal restriction fragment length polymorphisms of genes encoding 16S rRNA. Appl Environ Microbiol. 1997;63:4516-4522 35. Keuhl CJ, Wood HD, Marsh TL, et al. Colonization of the Cecal Mucosa by Helicobacter hepaticus Impacts the Diversity of the Indigenous Microbiota. Infect Immun. 2005;73:6952-6961 36. Abdo Z, Schuette UME, Bent SJ, et al. Statistical methods for characterizing diversity of microbial communities by analysis of terminal restriction fragment length polymorphisms of 16S rRNA genes. Environmental Microbiology. 2006;8:929-938 37. Martin FE, Nadkarni MA, Jacques NA, et al. Quantitative Microbiological Study of Human Carious Dentine by Culture and Real-Time PCR: Association of Anaerobes with Histopathological Changes in Chronic Pulpitis. J Clin Microbiol. 2002;40:1698-1704 38. Nitsche A, Becker M, Junghahn I, et al. Quantification of human cells in NOD/SCID mice by duplex real-time polymerase-chain reaction. Haematologica. 2001;86:693-699 39. Wirtz S, Neurath MF. Mouse models of inflammatory bowel disease. Adv Drug Deliv Rev. 2007;59:1073-1083 40. Erridge C, Duncan SH, Bereswill S, et al. The Induction of Colitis and Ileitis in Mice Is Associated with Marked Increases in Intestinal Concentrations of Stimulants of TLRs 2, 4, and 5. PLoS. 2010;5:e9125 41. Tang Y, Clayburgh DR, Mittal N, et al. Epithelial NF-{kappa}B Enhances Transmucosal Fluid Movement by Altering Tight Junction Protein Composition after T Cell Activation. Am J Pathol.176:158-167 114 42. Shea-Donohue T, Thomas K, Cody MJ, et al. Mice deficient in the CXCR2 ligand, CXCL1 (KC/GRO-{alpha}), exhibit increased susceptibility to dextran sodium sulfate (DSS)-induced colitis. Innate Immun. 2008;14:117-124 43. Blumberg RS, Saubermann LJ, Strober W. Animal models of mucosal inflammation and their relation to human inflammatory bowel disease. Curr Opin Immunol. 1999;11:648-656 44. Belardelli F, Ferrantini M. Cytokines as a link between innate and adaptive antitumor immunity. Trends Immunol. 2002;23:201-208 45. Abt MC, Artis D. The intestinal microbiota in health and disease: the influence of microbial products on immune cell homeostasis. Curr Opin Gastroenterol. 2009;25:496-502 46. Sartor RB. Microbial influences in inflammatory bowel diseases. Gastroenterol. 2008;134:577-594 47. Rath HC, Schultz M, Freitag R, et al. Different subsets of enteric bacteria induce and perpetuate experimental colitis in rats and mice. Infect Immun. 2001;69:2277-2285 48. Hudcovic T, Stepankova R, Cebra J, et al. The Role of Microflora in the Development of Intestinal Inflammation: Acute and Chronic Colitis Induced by Dextran Sulfate in Germ-Free and Conventionally Reared Immunocompetent and Immunodeficient Mice. Folia Microbiol. 2001;46:565-572 49. Kitajima S, Morimoto M, Sagara E, et al. Dextran sodium sulfate-induced colitis in germ-free IQI/Jic mice. Exp Anim. 2001;50:387-395 50. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, et al. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118:229-241 51. Heimesaat M, Fischer A, Siegmund B, et al. Shift towards proinflammatory intestinal bacteria aggravates acute murine colitis via Toll-like receptors 2 and 4. PLoS ONE. 2007;25:e662 115 52. Hoentjen F, Harmsen HJ, Braat H, et al. Antibiotics with a selective aerobic or anaerobic spectrum have different therapeutic activities in various regions of the colon in interleukin 10 gene deficient mice. Gut. 2003;52:17211727 53. Hansen R, Thomson JM, El-Omar EM, et al. The role of infection in the aetiology of inflammatory bowel disease. J Gastroenterol. 2010;45:266-276 54. Maukonen J, Satokari R, Matto J, et al. Prevalence and temporal stability of selected clostridial groups in irritable bowel syndrome in relation to predominant faecal bacteria. J Med Microbiol. 2006;55:625-633 55. Cummings JH, Macfarlane GT. Role of intestinal bacteria in nutrient metabolism. JPEN J Parenter Enteral Nutr. 1997;21:357-365 56. Araki Y, Mukaisho K, Sugihara H, et al. Proteus mirabilis sp. intestinal microflora grow in a dextran sulfate sodium-rich environment. Int J Mol Med. 2010;25:203-208 57. Wang M, Ahrne S, Jeppsson B, et al. Comparison of bacterial diversity along the human intestinal tract by direct cloning and sequencing of 16S rRNA genes. FEMS Microbiol Ecol; 2005:219-231 58. Derrien M, Vaughan EE, Plugge CM, et al. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol. 2004;54:1469-1476 59. Antonopoulos D, Huse S, Morrison H, et al. Reproducible community dynamics of the gastrointestinal microbiota following antibiotic perturbation. Infect Immun. 2009;77:2367-2375 60. Ivanov II, Frutos Rde L, Manel N, et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe. 2008;4:337-349 61. Ivanov II, Atarashi K, Manel N, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009;139:485-498 116 62. Ahmad MS, Krishnan S, Ramakrishna BS, et al. Butyrate and glucose metabolism by colonocytes in experimental colitis in mice. Gut. 2000;46:493-499 63. Barcenilla A, Pryde SE, Martin JC, et al. Phylogenetic Relationships of Butyrate-Producing Bacteria from the Human Gut. Appl Environ Microbiol. 2000;66:1654-1661 64. Arijs I, De Hertogh G, Lemaire K, et al. Mucosal gene expression of antimicrobial peptides in inflammatory bowel disease before and after first infliximab treatment. PLoS One. 2009;24:e7984 65. Wostmann B. The germfree animal in nutritional studies. Annu Rev Nutr. 1981;1: 257-279 66. Clarke TB, Davis KM, Lysenko ES, et al. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nature Medicine. 2010;16:228-231 67. Johansson ME, Phillipson M, Petersson J, et al. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. PNAS. 2008;105:15064-15069 68. Joerg DS, Svenja P, Maren A, et al. Epithelial Tight Junctions in Intestinal Inflammation. Ann NY Acad Sci. 2009;1165:294-300 69. Nenci A, Becker C, Wullaert A, et al. Epithelial NEMO links innate immunity to chronic intestinal inflammation. Nature. 2007;446:557 70. te Velde A, de Kort F, Sterrenburg E, et al. Comparative analysis of colonic gene expression of three experimental colitis models mimicking inflammatory bowel disease. Inflamm Bowel Dis. 2007;13:325-330 71. Podolsky D, Gerken G, Eyking A, et al. Colitis-associated variant of TLR2 causes impaired mucosal repair because of TFF3 deficiency. Gastroenterol. 2009;137:209-220 72. Yan Y, Kolachala V, Dalmasso G, et al. Temporal and spatial analysis of clinical and molecular parameters in dextran sodium sulfate induced colitis. PLOS One. 2009;4:e6073 117 73. Ito R, Kita M, Shin-Ya M, et al. Involvement of IL-17A in the pathogenesis of DSS-induced colitis in mice. Biochem Biophys Res Commun. 2008;377:12-16 74. Stefan L, Dick SD, Eva J, et al. The role of the Lps gene in experimental ulcerative colitis in mice. APMIS. 1996;104:823-833 75. Salzman NH, Bevins CL. Negative interactions with the microbiota: IBD. Adv Exp Med Biol. 2008;635:67-78 76. Rakoff-Nahoum S, Hao L, Medzhitov R. Role of Toll-like Receptors in Spontaneous Commensal-Dependent Colitis. Immun. 2006;25:319-329 77. Stanislawowski M, Wierzbicki PM, Golab A, et al. Decreased Toll-like receptor-5 (TLR-5) expression in the mucosa of ulcerative colitis patients. J Physiology Pharmacol. 2009;60:71-75 78. Sabine MI, Megan EH, Gijs H, et al. TLR5 is not required for flagellinmediated exacerbation of DSS colitis. Inflamm Bowel Dis.16:401-409 79. Gobert AP, Cheng Y, Akhtar M, et al. Protective Role of Arginase in a Mouse Model of Colitis. J Immunol. 2004;173:2109-2117 80. Horowitz S, Binion DG, Nelson VM, et al. Increased arginase activity and endothelial dysfunction in human inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1323-1336 81. Philip A, Nicholas CZ, Thuan N, et al. Distinct cytokine patterns identified from multiplex profiles of murine DSS and TNBS-induced colitis. Inflamm Bowel Dis. 2009;15:341-352 118 This chapter is in preparation for publication in Gut Microbes. Nagalingam NA, Robinson CJ, Young VB. The effects of intestinal microbial community structure on disease manifestation in IL10-/- mice infected with Helicobacter hepaticus. I performed experiments and processed samples for the cefoperazone study, and analyzed sequence data for both cefoperazone and vancomycin studies. Dr. Robinson conducted the vancomycin study and also processed these samples. I prepared the manuscript with contribution from two other authors. 119 Chapter 4 The effects of intestinal microbial community structure on disease manifestation in IL-10-/- mice infected with Helicobacter hepaticus. Abstract and Key Words: Backgroud: Inflammatory bowel disease (IBD) is an idiopathic disease where inflammation is associated with several factors, including changes in the intestinal microbiota. Here, we investigated the role of the microbiota in Helicobacter hepaticus infected IL-10-/- mice, a commonly used IBD model, in disease manifestation. We hypothesized that alterations in the microbiota affect disease severity during disease progression. Method: We altered the microbiota in mice by infecting animals with H. hepaticus and treating with vancomycin. We also infected animals with H. hepaticus after inducing alteration in the microbiota with cefoperazone. Through surveys of the 16S rRNA encoding-gene, analyses of histology and changes in host mediator expression, we measured changes in the microbiota and host response, Results: We found that severity of disease is independent of antibiotic-induced changes in the microbial community structure. However, we also documented differences in expression of host inflammatory mediators associated with shifts in the microbiota, though these differences led to the same pathology. 120 Conclusion: In conclusion, we demonstrated that disease is initiated and progresses in the presence of several different microbial communities. Also, H. hepaticus is the main driver of inflammation in this model, with changes in the microbiota playing a minor role in disease manifestation once inflammation is triggered. Finally, cefoperazone-induced changes in the microbiota may have long-term effects on the expression of host mediators. Key words: microbiota, IBD, Helicobacter hepaticus, IL-10-/- mice, 16S rRNA 121 Introduction: Inflammatory bowel disease (IBD) is most often represented by Crohn’s disease (CD) and ulcerative colitis (UC), but is a collective term referring to an idiopathic condition that results in inflammation of the intestinal mucosa. Although incidence of this disease is increasing (1, 2), its cause is yet to be determined. However, advances in the field point to genetic factors, dysregulated immune responses and altered intestinal microbial communities as playing key roles in the onset of disease (3). Dysbiosis, or alteration of microbial community structure, has been associated with intestinal diseases, both in patients and animal models of IBD (4-8). For example, fecal and biopsy samples from IBD patients have different microbial communities than those from healthy controls (9). Furthermore, patients with CD contain communities that are distinct from those in UC patients (9). Animal models have been key in elucidating the importance of not only the interaction between the immune system and the microbiota, but also the structure of the microbiota in IBD. Mice that are deficient in both Tbet, a transcriptional factor, and Rag2, recombination activating gene, contain colitogenic communities that can be transferred into healthy recipients and subsequently result in disease (10). Additionally, studies utilizing animals raised in germfree or gnotobiotic conditions provide further evidence for the necessity of the resident community and the significance of community structure in enteric diseases (11-13). The results of these studies suggest that changes in the balance among members of the indigenous microbial community can trigger an inflammatory response. 122 To further explore the role of the microbiota in IBD, several animal models are available used to monitor changes in these communities and their effects on disease, in a controlled environment (14, 15). A common model of IBD employs the infection of Helicobacter hepaticus in interleukin 10 (IL-10) deficient mice. In this model, infection elicits a Th1/Th17 response that most resembles Crohn’s disease in humans. Infection with H. hepaticus does not cause disease in wildtype mice and only triggers inflammation in immune altered hosts (16, 17). Onset of disease is MyD88 dependent (18). Additionally, H. hepaticus only causes disease in the presence of the resident microbiota with no disease observed in germfree mice (19), indicating the importance of the microbiota in the onset of IBD in this model. However, apart from these findings there is limited knowledge of the role that the resident microbiota plays in this model. Therefore, the aim of this study is to investigate the role of the microbiota in this model of IBD by altering community structure and observing disease manifestation. Previously we showed that treatment with cefoperazone significantly alters the microbial community of the murine gut (20), as does vancomycin (21). We used these antibiotics to modify the gut community structure in IL10-/- C57BL/6 mice before and during infection with H. hepaticus, and then monitored disease. We hypothesized that the structure of the microbiota is related to disease severity in IL-10 deficient mice infected with H. hepaticus. 123 Materials and methods: Animals: C57BL/6 IL-10-/- mice from a breeding colony initially established with breeding stock from Jackson Laboratories (Bar Harbor, ME) were use for all experiments. Mice were housed with autoclaved bedding, given sterile food and water ad libitum, and exposed to 12:12 h light:dark cycles. Germfree mice were housed in sterile microisolators. All animal studies were conducted at the University of Michigan and were approved by the University Committee on Use and Care of Animals (UCUCA). Murine infection with H. hepaticus. The wild-type H. hepaticus strain 3B1 (type strain, ATCC 51488) was incubated at 37!°C under microaerobic conditions for 48 h, on Tryptic Soy Agar plates (TSA), supplemented with 5% sheep’s blood, and then re-suspended in 5 ml of tryptic soy broth (TSB). Mice were inoculated with a suspension of bacteria with an optical density of 1.0 at 600 nm (~108 CFU) in a volume of 0.2 to 0.3 ml. Bacteria were introduced directly into the stomach with a 24-gauge ball-tipped gavage needle. Control mice were inoculated with sterile TSB. Three experimental studies were conducted to investigate roles of the microbiota in disease development: 1) To investigate whether disease can develop in H. hepaticus monoassociated IL-10 deficient C57BL/6 mice in our colonies, two groups of germfree mice were used: five mice were gavaged with TSB only and five were gavaged with H. hepaticus. Tissues were collected four weeks postinfection. 2) To investigate roles of an altered microbiota in disease development in this model, we changed microbial communities using two different antibiotic 124 treatments. Cefoperazone was administered, ad libitum, as a 0.5 mg/ml solution in the drinking water of 10 weeks old, conventionally raised C57Bl/6 IL-10 deficient mice. Mice were assigned to six groups, as shown in figure 4.1A: one group was infected with H. hepaticus after 1 day recovery from cefoperazone, another group infected after 6 weeks recovery from cefoperazone, and a third group infected without prior cefoperazone treatment. Three other groups were used as the uninfected controls for respective cefoperazone treatments 3) The second antibiotic treatment involved ad libitum administration of 0.1 mg/ml vancomycin in the drinking water of 6-8 week old, conventionally-raised C57Bl/6 IL-10 deficient mice (21). Mice were assigned to six experimental groups as depicted in figure 4.1B: One group of mice was given vancomycin then infected with H. hepaticus as antibiotic administration continued. A second group had similar treatment but vancomycin treatment was ceased 10 days post infection as mice recovered fro 11 days from antibiotic treatment. A third group of mice was used as vencomycin untreated controls. Three H. hepaticus uninfected control groups were used for respective antibiotic treatments. Mice were grouped into cages according to gender and treatment. 125 A C57BL/6 IL10-/- mice (n=36) Cef 10 days (n=12) Control (n=14) Hh uninfected 4 weeks (n=7) Hh 4 weeks (n=7) Hh uninfected 4 weeks (n=7) Cef 10 days (n=10) 1 day off drug 6 weeks off drug Hh 4 weeks (n=5) Hh uninfected 4 weeks (n=5) Hh 4 weeks (n=5) Harvest cecum for microbial ecology analyses B C57BL/6 IL10-/- mice (n=48) Vancomycin 14 days (n=34) Control (n=14) Hh uninfected 10 days (n=6) Hh uninfected 10 days (n=6) Hh10 days (n=8) Hh uninfected 10 days (n=6) Hh 10 days (n=10) Hh 10 days (n=12) 11 days off drug Harvest cecum for microbial ecology analyses Figure 4.1 Schematic of experimental designs showing (A) cefoperazone (Cef) and (B) vancomycin administration to IL10-/- C57BL/6 mice prior to or during infection with H. hepaticus (Hh). 126 Necropsy and Histology: Mice were euthanized by CO2 asphyxiation and tissues harvested (22). The cecum sections were gently washed with 1X phosphate-buffered saline (PBS) to remove luminal contents, cut into sections and snap-frozen in liquid nitrogen and stored at -80oC until future use. The remaining portion of the cecum was processed for histology by removing the luminal contents before washing with PBS, and placing the tissue in tissue cassettes. Cassettes were then submerged in 10% formalin for 24 hours and transferred to a 60% ethanol solution prior to processing for paraffin embedding and staining with hematoxylin and eosin (H&E). Histologic sections were scored by a veterinary pathologist who was blind to the sample source. The following scoring system was used: 0, no significant inflammation; 1, small multifocal lamina proprial or transepithelial leukocyte aggregates; 2, coalescing lamina proprial leukocyte aggregates, may have submucosal involvement; 3, frequent coalescing leukocyte aggregates with submucosal involvement, may have follicle formation; 3.5, strong submucosal component, infrequent extension to muscularis externa or mesocolon; 4, diffuse or regionally extensive transmural involvement. DNA extraction: Genomic DNA was extracted from tissue using the MagNa Pur® system (Roche, Indianapolis, IN). Briefly, the tissue was incubated in lysis buffer and proteinase K at 56 oC, before being placed in the MagNa Pur® Compact machine for purification of DNA. The resulting DNA was then used in preparation of the clone libraries and quantitative PCR (qPCR). 127 Clone library construction: PCRs were set-up as described in Young et al. (23). Briefly, illustra PuReTaqTM Ready To Go PCRTM beads (GE Healthcare, Piscataway, NJ). and AGAGTTTGATCCTGGCTCAG-3'; broad-ranged 1492R, primers, (8F, 5'- 5'-GGTTACCTTGTTACGACTT-3') were used to amplify 16S rRNA genes. GE illustraTM MicrospinTM S-400 HR columns were used to purify amplicons (GFX, GE Healthcare, Piscataway, NJ) as directed by the manufacturer. Purified PCR products were ligated into the TOPO 4® vector (Invitrogen K4575-01, Carlsbad, CA) according to manufacturer’s specifications, and transformed into Escherichia coli TOP10 cells. Clones were grown overnight at 37 oC in Luria Broth (LB) amended with carbenicillin (50 µg/ml)). Vector CAGTCACGACGTTGTAAAACGACGGC-3'; specific and primers (M13F, M13R, 5'5'- CAGGAAACAGCTATGACCATG-3') were used to screen for clones containing the appropriately sized inserts and to amplify inserts for sequencing. Partial 16S rRNA gene sequences were generated by sequencing with primer 8F, at the Sequencing Core at the University of Michigan. Raw sequence data were processed through an automated “information pipeline” available through the Ribosomal Database Project (RDP) Web site (http://rdp.cme.msu.edu/). Distance matrices containing data from each library were calculated from alignments generated by the RDP aligner (24). The program mothur (25) was used to assign sequences to operational taxonomic units (OTUs) based on values in the distance matrices. Analyses were done using a 95% sequence similarity to denote genus level and at a 80% sequence similarity to approximate phylum 128 level (25). EstimateS (26) and mothur were used to calculate ecological measures from the distance matrices. The Morisita Horn index was then used to measure the similarity among samples taking into account both abundance and richness of the community. Phylogenetic trees were calculated using the Mega3 program and the parsimony test from the mothur suite of programs was used to denote significant differences between treatment groups (25, 27). Additionally, taxonomic assignments were determined using Classifier through the RDP website (80% confidence cut-off). qPCR analysis of microbial communities: The quantity of 16S rRNA operons in the samples relative to a single-copy host gene was measured using a primer/probe set that targets a broad range of rRNA-encoding gene sequences (forward primer (5’-TCCTACGGGAGGCAGCAGT-3’); GGACTACCAGGGTATCTAATCCTGTT-3’); and reverse probe primer (5’- (5’-[6-FAM]- CGTATTACCGCGGCTGCTGGCAC-[TAMRA]-3’)) (28). A primer/probe set targeting a 264-bp portion of the TNF alpha gene was used as a reference using 200 nanomoles of the forward (TNFα_mu_se; 5’- GGCTTTCCGAATTCACTGGAG-3’) and reverse primers (TNFα_mu_as; 5’CCCCGGCCTTCCAAATAAA-3’), (TNFα_mu_probe; and 100 nanomoles of the probe 5’-[Cy5]-ATGTCCATTCCTGAGTTCTGCAAAGGGA-[Iowa Black RQ™]-3’) (29). The reaction mix consisted of LightCycler® 480 Probes Master reaction mix (Roche) at 1X concentration, and appropriate primer/probe pair. Amplification of each gene was done under separate run conditions: the 16S rRNA gene target had an activation step of 50 oC for 2 min followed by 95 oC 129 for 10 min. Forty-five cycles were done at 95 oC for 15 s and 60 oC for 1 min before holding at 4 oC. The TNF reference gene was also amplified with an activation step of 50 oC for 2 min followed by 95 oC for 10 min. Forty-fives cycles of 95 oC for 20 s and 64 oC for 30 s were done before holding at 4 oC. Calculations of 2-ΔΔCt were made to compare changes in the amount of 16S from samples between treatment groups (21,22). RNA extraction and determination for cytokine expression: Total nucleic acids were extracted by MagNaPur® (Roche) as directed by the manufacturer’s protocol. Total RNA was then isolated by treating total nucleic acids with DNaseI recombinant, RNase-free (Roche, Indianapolis, IN). The High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, CA) was used to transcribe total RNA to cDNA. Cytokine expression was determined by applying the cDNA to a custom Superarray® (Frederick, MD) PCR plate which contained gene-specific primers designed for the following targets (30): IFNγ, IL-12a, TNFα, IL-4, IL-13, IL-5, TGFβ, IL-10, IL-6, FOXp3, IL-17a, IL-23a, CXCL2, CCL5, NOS2, Arg1, Chi3/4, PTGS2, INDO, GAPDH, βActin, a reverse-transcription control and a genomic DNA control. Quantization was done with LightCycler® 480 SYBR Green I Master and analyzed on a LightCycler® 480 system (Roche Diagnostics, Indianapolis, IN) as directed by manufacture’s protocol, with the following program: 95 oC activation for 10 min; 40 cycles of 95 oC denaturation for 15s, and 60 oC annealing for 1 min. Resulting threshold values were analyzed by calculating the 2-ΔΔCt values, using GAPDH as the reference, to calculate fold 130 regulation compared to the antibiotic untreated, H. hepaticus uninfected control (31-33). Statistical Analysis: Statistical significances were determined using KruskalWallis tests for the histological scores, and ANOVA tables for the microbial abundances among the treatment groups using the Metastats, software for metagenomic analysis (34). The differences in the dendrograms were calculated using the parsimony test function in mothur (25). Probability values less than 0.05 were considered significantly different. Results: Germfree mice infected with H. hepaticus do not get colitis. To investigate whether H. hepaticus can colonize germfree mice from our colonies, we infected IL-10 deficient animals and monitored them for 4 weeks. Absence of disease when mice are monoassociated with H. hepaticus would indicate that the resident microbiota has an important role in disease manifestation. Detection of H. hepaticus was done through qPCR, and revealed that the loads in germfree mice were 2.94 ±1.90 fold less than loads in conventional mice, a level that was not significantly different (p=0.446) (table 4.1). This indicates that not only can H. hepaticus colonize germfree mice, as indicated by others (19), but that the bacterium may have a particular niche that is unaffected by the presence of resident microbes. 131 Treatment Cefoperazone treatment No cef 1 day recovery from cef 1 day recovery from cef before infection No cef 1 day recovery from cef before infection 6 wk recovery from cef before infection 6 wk recovery from cef before infection Vancomycin treatment No vanc Vanc No vanc Vanc Vanc + 11 days recovery Vanc + 11 days recovery H. hepaticus infection No No Fold change ± SD Total bacterial H. hepaticus load calibrator N/A -3333.00 ± 0.34 N/A No 2.16 ± 1.18 N/A Yes 1.25 ± 0.75 calibrator Yes 4.48 ± 2.55 -1.12 ± 2.79 Yes -1.35 ± 0.71 3.94 ± 6.30 No 1.14 ± 0.04 N/A No No Yes Yes Yes No 1.17 ± 0.68 1.95 ± 1.41 1.90 ± 1.91 1.98 ± 1.62 1.14 ± 1.16 1.41 ± 0.91 N/A N/A 1.04 ± 0.30 -1.59 ± 0.52 1.28 ± 0.56 N/A N/A -2.94 ± 1.90 Germfree mice infected with H. hepaticus Table 4.1. Quantitative PCR measures of fold changes in the total bacterial and in H. hepaticus loads from genomic DNA extracted from cecum tissue. The fold change reported (2-ΔΔCt) is normalized to GAPDH and in comparison to the samples indicated as “calibrator” in the respective columns. 132 Through histological analysis, no inflammation was detected in cecum tissues from germfree mice that were infected with H. hepaticus (figure 4.2). This result is in agreement with a previous study (19). However, unlike previous reports (19), IL-10 deficient mice raised in specific pathogen-free conditions did not develop inflammation. This indicates that spontaneous disease is only associated with mice containing specific microbial communities, again emphasizing the important role of the microbiota in disease development. 133 A B E C D F 50 m ** Inflammation Score E Mouse H. hepaticus 4 3 ns 2 1 0 Germfree Germree uninfected infected Conv Conv uninfected infected Figure 4.2 Helicobacter hepaticus triggered inflammation in conventionally raised IL10-/- C57BL/6 mice compared to germfree counterparts. Representative sections of mouse cecum from experimental groups are depicted. (A) H. hepaticus uninfected conventionally raised mice appear normal. (B) H. hepaticus infected conventional (Conv) section shows edema, hyperplasia and inflammatory infiltrate. (C) H. hepaticus uninfected germfree control, and (D) H. hepaticus infected germfree cecum sections are both normal without signs of inflammation. (E) H. hepaticus forms close association to the intestinal epithelium, and in the crypts (yellow) compared to other microbes (red) in the lumen. (F) In germfree mice monoassociated with H. hepaticus, the bacteria are also found in close association with the epithelial layer (G) Quantification of indicators of pathology showing significant inflammation only in H. hepaticus infected conventionally raised mouse sections. Asterisk (*) denotes statistically significant changes (p<0.05). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. 134 Vancomycin alters communities in uninfected mice. Vancomycin was administered to mice infected with H. hepaticus prior to infection in order to investigate the effects of altering community structure on the development of inflammation. Through clone library construction, we found that vancomycin treated communities were significantly altered and these changes persisted even after administration ceased. Although bacterial load, via qPCR, was not altered by vancomycin treatment (table 4.1), this antibiotic caused a decrease in overall microbial diversity and an increase in relative abundance of members of the Proteobacteria, namely E.coli/Shigella (figure 4.3). Also, total community richness was reduced in vancomycin treated mice (Shannon index= 2.10± 0.55) compared to untreated controls (Shannon index= 3.55± 0.15) (supplemental figure 4.3). These results indicate that vancomycin alone causes changes in the microbial community. Communities did not cluster according to gender or housing. 135 Changes in vancomycin treated communities infected with H. hepaticus * vancomycin * H. hepaticus 0.4 0.3 0.2 0.1 0.0 Anaeroplasma Akkermansia Escherichia/Shigella Helicobacter Barnesiella unclassif Porphyromonadaceae Clostridium Dorea Coprococcus unclassif Lachnospiraceae Anaerotruncus Oscillibacter Ruminococcus Butyricicoccus unclassif Ruminococcacea Peptococcus unclassified Peptococcaceae Anaerovorax unclassif Clostridiales Allobaculum Holdemania Coprobacillus Turicibacter unclassif Erysipelotrichaeae Lactobacillus NA Control Recovered from vancomycin Vancomycin control + H. hepaticus Recovered from vancmycin + H. hepaticus Vancomycin + H. hepaticus Figure 4.3 H. hepaticus infection, as well as vancomycin treatment, significantly alters intestinal community structure. Asterisk (*) denotes statistically significantly different community structures calculated by parsimony test (p<0.05). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. 136 Vancomycin and H. hepaticus alter communities in infected mice. Investigations on the effects of H. hepaticus infection on the microbial community structure, during vancomycin treatment, showed reduction in community richness (Shannon index= 1.35± 0.35) compared to microbiota in infected, vancomycinuntreated mice (Shannon index= 2.47± 1.01) (supplemental figure 4.3). We also found that mice that were continuously administered vancomycin and infected with H. hepaticus (VancHh) contained the same load of H. hepaticus as mice that were not administered antibiotics (Hh) (p=0.06). However, VancHh mice contained fewer H. hepaticus than mice that had been administered vancomycin previous to H. hepaticus infection, but not during infection (VancRecHh) (p=0.009). There was no difference between the loads in Hh and VancRecHh mice (p=0.28). Structurally, the communities in VancRecHh mice were more like the communities in mice that were only infected with H. hepaticus and no antibiotic (Hh) (Morisita-Horn distance= 0.36) than those in VancHh mice (Morisita-Horn distance= 0.43). As compared to the communities in VancRecHh mice, VancHh mice contained communities with decreased population sizes of Oscillibacter (p=0.005), Lachnospiraceae (p=0.003), Coprocococcus (0.011) and Dorea (p=0.038). Additionally, the VancHh communities contained more E. coli/Shigella (p=0.001), Akkermansia (p=0.001) and Anaeroplasma (p<0.001). Shannon diversity index indicates that communities in VancRecHh mice are more diverse than communities in VancHh mice (p= 0.001). Taxonomic analyses indicated that communities in VancRecHh also exhibited 137 a significant increase in Lachnospiraceae (p=0.001) and decrease in E. coli/Shigella (p=0.005) as compared to communities in VancHh mice (figure 4.3). Inflammation occurs in vancomycin treated mice infected with H. hepaticus. To investigate whether changes in the microbiota affect disease manifestation, cecum sections of vancomycin treated mice were scored. All animals infected with H. hepaticus developed inflammation, regardless of the shifts in the microbial community structures (figure 4.4). H. hepaticus infection alters communities in infected mice. Since vancomycin treatment did not result in differences in disease manifestation in H. hepaticus infected mice, we administered cefoperazone to animals since it is known to cause significant, broad-spectrum changes to the intestinal microbiota. The community 1 day following cefoperazone treatment has over a 1000 fold lower bacterial load (table 4.1). After cefoperazone treatment, mice were administered untreated water and allowed to recover for one day, or for six weeks before infecting with H. hepaticus. Four weeks post infection, all communities had similar total bacterial loads compared to the cefoperazone untreated mice that were H. hepaticus uninfected, indicating that the communities had recovered during the four weeks. This was also supported by diversity measures (supplemental figure 4.3). Communities in infected mice contained similar amounts of H. hepaticus regardless of cefoperazone treatment or recovery period (table 4.1). It is 138 noteworthy that there was an exception, with one infected mouse having 20-fold less detectable CdtB target, indicating a low H. hepaticus load. 139 * Inflammation Score 4 3 2 1 0 no yes yes 11 va nc + va nc + yes va da nc ys re co ve ry va da nc ys re co ve ry No ne no 11 Vancomycin treatment no No ne H. hepaticus infection Figure 4.4 Blinded scores showing significant inflammation in all H. hepaticus infected sections regardless of vancomycin treatment compared to uninfected controls. Asterisk (*) denotes statistically significant changes (p<0.05). 140 Clone libraries were built from three individual mice from each treatment group (supplemental 4.1). OTU-based analyses show that the communities in H. hepaticus-infected mice were significantly different from those in mice that did not harbor H. hepaticus (p<0.001). In communities of infected mice, the relative abundance of H. hepaticus varied between 25% and 70% (figure 4.5). This difference in relative abundance of H. hepaticus was independent of cefoperazone treatment. The mouse that contained low numbers of H. hepaticus, as determined via qPCR, contained a community that produced only 2 of 90 clones that affiliated with Helicobacter. This supports the finding that H. hepaticus did not dominate this community as was typically seen in the other mice in this treatment group. There were reductions in the genera Clostridia (p=0.032) and family Lachnospiraceae (p=0.016) in communities infected with H. hepaticus after only one day off cefoperazone compared to those infected after six-weeks recovery. This indicates differences in recovery dynamics in the two treatment groups. There were no gender or housing effects on communities’ relatedness. 141 Changes in cefoperazone treated communities infected with H. hepaticus * H. hepaticus 0.4 0.3 0.2 0.1 0.0 Anaeroplasma Akkermansia Escherichia/Shigella Helicobacter Barnesiella unclassif Porphyromonadaceae Clostridium Dorea Coprococcus unclassif Lachnospiraceae Anaerotruncus Oscillibacter Ruminococcus Butyricicoccus unclassif Ruminococcacea Peptococcus unclassified Peptococcaceae Anaerovorax unclassif Clostridiales Allobaculum Holdemania Coprobacillus Turicibacter unclassif Erysipelotrichaeae Lactobacillus NA Control Recovered from cefoperazone for 1 day Recovered from cefoperazone for 6wks control + H. hepaticus Recovered from cefoprazone 1 day + H. hepaticus Recovered from cefoprazone 6 wks + H. hepaticus Figure 4.5 H. hepaticus infection significantly alters the community structure of mice even after four weeks of termination of cefoperazone treatment. Asterisk (*) denotes statistically significantly different community structures calculated by parsimony test (p<0.05). For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. 142 Inflammation occurs in cefoperazone treated mice infected with H. hepaticus. To investigate whether a major perturbation would affect disease manifestation in H. hepaticus infected IL-10 deficient mice, cefoperazone was administered to mice before infecting with the bacterium. Cecum sections were then scored, and it was found that all animals infected with H. hepaticus developed inflammation (figure 4.6). There was no correlation between disease scores and relative abundance of H. hepaticus (Spearman’s ρ = 0.242). Despite changes to the microbiota, cefoperazone did not alter disease severity in H. hepaticus infected mice as compared to infected mice that were not administered antibiotics. 143 * 2 1 no yes ce f co v w ks re v y da 1 no 6 re co re c ks w 6 no ce f ne no ov ce f ov re c 1 H. hepaticus infection ce f 0 da y Cefoperazone treatment prior to infection 3 no ne Inflammation Score 4 yes yes Figure 4. 6 Blinded scores showing significant inflammation in all H. hepaticus infected sections regardless of cefoperazone treatment compared to uninfected controls. Asterisk (*) denotes statistically significant changes (p<0.05). 144 Infection with H. hepaticus and/or cefoperazone induces host response. Although development of inflammation was not affected by cefoperazone treatment, qPCR showed that antibiotic treated mice that were infected 1 day after treatment ceased, had a lower load of H. hepaticus than animals that were allowed to recover from treatment for six weeks prior to infection. To investigate possible host effects due to this difference, we measure changes in expression of host mediators. This analysis indicated that changes were elicited by administration of cefoperazone in communities containing H. hepaticus. After cefoperazone treatment, there was a significant up-regulation of several host mediators including TNFα, CCL5, CCL2, Nos2 and Arg1, or down-regulation in CHI3/4 and IL-23a (figure 4.7, supplemental 4.2). Even when mice were allowed to recover for 6 weeks from cefoperazone treatment, expression of some mediators remained significantly different from the no-cefoperazone, H. hepaticus infected control, indicating that treatment with cefoperazone causes an altered immune response in the presence of this bacterium. These changes do not mediate lesions. 145 * Fold regulation compared to no cefoperazone, H. hepaticus infected control 40 Cefoperazone 1 day recovery, H. hepaticus Cefoperazone 6 wk recovery, H. hepaticus 25 20 * 15 * 10 * 5 IL-23a 0 -5 * * * CHI3/4 * * -10 IL-12a -15 * * TGFb TNF * * CCL5 CCL2 -50 NOS2 ARG1 INDO PTGS2 * Figure 4.7 Changes in expression of host mediators in cecum tissue of mice administered cefoperazone and infected with H. hepaticus (p<0.05). 146 Discussion: Intestinal microbes have been credited with many beneficial functions including break-down of complex carbohydrates, protection against pathogens and education of the immune system (35). These are important interactions between the host and its resident microbes as seen by deficiencies in the host development in animals raised in germfree conditions (36-38). Collapse of this delicate balance has been implicated in the onset of diseases, such as inflammatory bowel disease (IBD), a condition triggered by an inappropriate response by the hosts’ own immune system. It is also associated with an altered microbiota, a condition commonly referred to as dysbiosis. Patients with IBD often exhibit a reduction in diversity of the microbial community structure compared to healthy participants (39-42). Additionally, some investigators noted a relative increase in particular bacterial groups such as the Bacteriodetes in patients with IBD compared to healthy counterparts (43, 44). There is also evidence that the microbial community is important in the persistence of disease with antibiotic studies showing amelioration of IBD after treatment (45, 46). Apart from this we have little insight into the role of the microbiota in IBD. We therefore used a murine model of IBD, Helicobacter hepaticus infected IL-10 deficient C57BL/6 mice, to further investigate how changes in the microbial community associated with disease by testing whether the severity of disease is affected by different microbial community structures. Helicobacter hepaticus is sometimes found as part of the resident intestinal community of mice (47-49), and triggers inflammation in immuno-compromised 147 mice such as Rag-/-, IL-10-/- and SCID (50-52). Therefore, this bacterium is a pathobiont, a member of the resident bacteria with the potential to cause disease (53). Furthermore, H. hepaticus-induced inflammation can be ameliorated by the use of antibiotics (54, 55). However, Helicobacter causes disease in IL-10 deficient mice only in the presence of the resident microbiota indicating an essential role of the indigenous community towards inducing inflammation in this model (19). Although other groups have shown the development of spontaneous disease in IL-10 deficient mice (11, 19), we report less than 5% of mice in our breeding colony develop disease over a 2 year period (17), suggesting that disease is associated with certain microbial communities. These data collectively suggest that the microbiota is an important driver of inflammation in IBD, particularly in this model, making it an appropriate system in which to study the effects of an altered microbial community structure on severity of inflammation. To investigate the extent of the role of the microbiota, we infected mice containing various communities with H. hepaticus and assessed whether there were differences in disease severity. Two antibiotics, vancomycin and cefoperazone, were used to alter the microbial communities. Each drug causes different changes in the gut community: vancomycin, a drug that targets the gram-positive bacteria, caused shifts in the microbial structure without reducing the bacterial load, and cefoperazone, a broad-spectrum antibiotic, reduced the total bacterial load almost 1000-fold as we have shown here and previously (20, 21). With vancomycin, these shifts were mainly due to the reduction in the Firmicutes, a phylum that typically comprises more than 50% of the gut 148 community (23, 56). After a recovery period communities exposed to vancomycin began to resemble control communities, but also exhibited lasting effects with a reduction of the phylum Firmicutes and an increase in the Proteobacteria. Increases in the Proteobacteria after vancomycin treatment have also been observed by other investigators (57), and are perhaps due to the reduction in gram-positive bacteria, thereby making formerlly occupied niches available to members of the Proteobacteria. Although we did not detect them in control groups in this study, previously we showed that members of the Proteobacteria inhabit the ceca of IL-10-/- mice using 454 pyrosequencing analysis of the microbiota (20). It is likely that they were present in the communities analyzed in the current study, but were below the detection limits of clone library construction. Like other communities (23, 58) after a recovery period, cefoperazone treated communities began to resemble the communities in untreated controls, demonstrating resilience, even after a significant reduction in bacterial loads immediately after treatment. Perturbation of the microbiota can enhance colonization by pathogens as seen with increased loads of Salmonella in mice treated with antibiotics (57, 59-61). In this study, cefoperazone and vancomycin treatments caused different changes in the microbiota, and although the relative proportions of the H. hepaticus were different among the various communities, the bacterium’s loads were comparable to that in the antibiotic-untreated group. These data suggest that H. hepaticus may have a specialized niche as its colonization is unaffected by the alterations in these resident microbiota. One study shows that Salmonella exhibits similar 149 colonization abundance when introduced into mice containing two different microbial communities, one perturbed by multi-drug cocktail and another that had recovered (61). The observation suggested to investigators that since the communities were unlikely to be restricting the Salmonella colonization, it is plausible that host innate immune system was enforcing the resistance (61). Future ecological studies can contribute to understanding the mechanism by which H. hepaticus interacts with the microbiota in induction of disease. From the histological analyses, H. hepaticus infection caused infiltration by inflammatory cells indicative of IBD (62). Disease occurrence was independent of community structure with the only common significant association, between disease and community change, being the presence of H. hepaticus. Even then, the relative abundance of this bacterium did not correlate with disease score. This finding also resonates with the outcome that severe disease occurs even at very low colonization loads of H. hepaticus, just as it does with relatively high colonization levels. Many factors contribute to the development of IBD (3), however these contributions may not be equal. There may be a skew towards one factor being a predominant contributor to disease development as seen in monozygotic twin studies where environment plays an important part, while genetics’ role is to a lesser degree (63); or where fecal transplants or probiotics lead to the amelioration of disease demonstrating an important role of the microbiota over other factors (64, 65). Likewise, this model indicates that the mere presence of H. hepaticus is a strong trigger for inflammation and changes in the resident community plays a comparatively minor role, if any. 150 The host immune response to H. hepaticus, such as increase in arginase and Nos2, was consistent with the work of others which revealed increases in iNOS (66) These enzymes are important in wound healing and are associated with increases in neutrophil activity, which was observed here. Alterations in the microbial community by antibiotic treatment have been associated with altered immune responses (67, 68), and since cefoperazone treatment is known to significantly reduce bacterial load in the intestine, it was surprising to observe that final inflammation exhibited was unaffected by antibiotic treatment. We decided to further investigate whether changes in other host response were altered by the treatment regime. We discovered that after recovery from cefoperazone treatment, and H. hepaticus infection, TNFα increased significantly while INFγ decreased, which may be a compensatory act for the lack of INFγ response in IL-10 deficient mice (69). Also, the decrease in chitinase (chi3/4) expression in cefoperazone treated mice may be a response triggered by the decrease in microbial load after antibiotic treatment. Chitinase is highly expressed in chemically-induced IBD and has a role in enhanced bacterial adhesion (70). With lower bacterial loads, chitinase expression may also be reduced through lack of microbial stimulation. Likewise, the increases in certain mediators such as TNFα and chemokines CCL5 and CCL2 seen in the cefoperazone treated mice, compared to untreated mice infected with H. hepaticus may be due to changes in the microbial stimuli due to shifts in the microbiota after antibiotic treatment (71). These findings not only demonstrate that cefoperazone treatment can have long-term effects on host response, but 151 that different expression of host mediators can result in similar disease manifestation in H. hepaticus infected mice. Perhaps the most significant finding of this work was that disease can be initiated in several different microbial communities. Also, after initiation, disease progressed to exhibit similar manifestations suggesting that microbiota structure may not be a strong factor in exacerbating or reducing disease in this model. The actual mechanism whereby the Helicobacter interacts with the microbiota during inflammation is yet to be determined. It is possible that the resident microbes prime the host immune system so that a response can be generated to the Helicobacter, or cross-talk with other microbes are needed for the H. hepaticus to trigger a host response. The microbiota may contribute to disease in multiple ways, making studies of potential patterns in the community important in unraveling the complexities of IBD perhaps leading to novel therapies or the development of prognostic tools for early detection of this debilitating condition. Acknowledgements I would like to extend thanks to Judith Opp and Kristen Frey for technical assistance, and to laboratory colleagues and Dr. Gary Huffnagle for advice and discussion of the manuscript. We would also like to especially thank the NIH for funding. 152 SUPPLEMENTAL INFORMATION Table 4.2. Clones generated from genomic DNA extracted from cecum samples. Treatment Cefoperazone treatment No cef 1 day recovery from cef before infection No cef 1 day recovery from cef before infection 6 wk recovery from cef before infection 6 wk recovery from cef before infection Vancomycin treatment No Vanc No Vanc Vanc + 11 days recovery Vanc + 11 days recovery H. hepaticus infection No No Clones/sample (N) Yes Yes 83, 92, 86 (3) 150, 163, 76 (3) Yes 93, 72, 58 (3) 93, 54, 93 (3) 90, 75, 92 (3) No No No Yes Yes Yes No 153 95, 95, 95 (3) 94, 84, 94 (3) 89, 89, 92, 93, 94 (5) 94, 95, 93, 90, 92, 94 (6) 94, 91, 92, 93, 91, 90, 96 (7) 95, 93, 92 (3) Table 4.3. Fold change in expressions of host mediators from cecum tissue. All values are normalized to GAPDH expression and compared to cefoperazone untreated, H. hepaticus uninfected control group. Asterisk (*) denotes statistically significant changes (p<0.05). 1 day recovery from cef, 6 wk recovery from cef, H. hepaticus H. hepaticus Host mediator IFNγ 1.94 ± 1.12 -2.70 ± 1.09 1.50 ± 1.57 * -3.85 ± 1.53 * IL-12a TNFα 1.06 ± 1.03 * 8.84 ± 1.50 * 1.77 ± 1.53 -1.85 ± 0.62 IL-4 -1.37 ± 1.21 -1.03 ± 1.97 IL-13 1.77 ± 1.53 -1.85 ± 0.62 IL-5 TGFβ -2.86 ± 0.68 * 4.77 ± 3.14 * 1.77 ± 1.53 -1.85 ± 0.62 IL-6 1.77 ± 1.53 -1.85 ± 0.62 FOXp 1.77 ± 1.53 -1.85 ± 0.62 IL-17a -1.52 ± 1.29 * -6.67 ± 1.01 * IL-23a 1.35 ± 1.85 -2.44 ± 0.94 CXCL2 1.59 ± 0.61 * 9.42 ± 0.56 * CCL5 -1.19 ± 1.18 * 25.00 ± 1.04 * CCL2 9.36 ± 2.24 * 41.5 ± 1.32 * NOS2 6.57 ± 3.16 * 4.17 ± 6.28 * ARG1 -7.69 ± 1.39 * -50.00 ± 1.05 * CHI314 2.38 ± 2.29 * -5.56 ± 1.25 * PTGS2 6.74 ± 1.82 * 3.26 ± 3.12 * INDO 154 Rarefaction curves (at 95% sequence similarity) showing decrease in diversity of microbial communities treated with vancomycin A 90 80 H. hepaticus uninfected OTU 70 60 50 40 30 20 10 0 0 B80 50 100 150 200 250 300 H. hepaticus infected 70 60 OTU 50 40 30 20 10 0 0 50 100 150 200 250 300 350 400 450 500 collection effort/sequences vancomycin control cefoperzone control vancomycin vancomycin recovered cefoperazone 4 wks recovered cefoperazone 10 wks recovered Figure 4. 8 (A) Rarefaction curves show a reduction in richness in communities treated with vancomycin, with or without the presence of H. hepaticus. (B) Communities recovering in the presence of H. hepaticus have lower community richness than communities in mice infected with H. hepaticus after 6 weeks off cefoperazone, or those in mice that were not administered cefoperazone. For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. 155 REFERENCES 156 REFERENCES 1. Lakatos P. Recent trends in the epidemiology of inflammatory bowel diseases: up or down? World J Gastroenterol. 2006;12:6102-6108 2. Gismera CS, Aladren B. Inflammatory bowel diseases: a disease (s) of modern times? Is incidence still increasing? World J Gastroenterol. 2008;4:54915494 3. Packey C, Sartor R. Interplay of commensal and pathogenic bacteria, genetic mutations, and immunoregulatory defects in the pathogenesis of inflammatory bowel diseases. J Intern Med. 2008;263:597-606 4. Chang JY, Antonopoulos DA, Kalra A, et al. Decreased Diversity of the Fecal Microbiome in Recurrent Clostridium difficile-Associated Diarrhea. J Infect Dis. 2008;197:435 5. Swidsinski A, Weber J, Loening-Baucke V, et al. Spatial Organization and Composition of the Mucosal Flora in Patients with Inflammatory Bowel Disease. J Clin Microbiol. 2005;43:3380-3389 6. Thompson-Chagoyan OC, Maldonado J, Gil A. Aetiology of inflammatory bowel disease (IBD): Role of intestinal microbiota and gut-associated lymphoid tissue immune response. Clin Nutri. 2005;24:339-352 7. Araki Y, Andoh A, Tsujikawa T, et al. Alterations in intestinal microflora, faecal bile acids and short chain fatty acids in dextran sulphate sodium-induced experimental acute colitis in rats. Eur J Gastroenterol Hepatol. 2001;13:107-112 8. Heimesaat M, Fischer A, Siegmund B, et al. Shift towards proinflammatory intestinal bacteria aggravates acute murine colitis via Toll-like receptors 2 and 4. PLoS ONE. 2007;25:e662 9. Swidsinski A, Loening-Baucke V, Vaneechoutte M, et al. Active Crohn's disease and ulcerative colitis can be specifically diagnosed and monitored based on the biostructure of the fecal flora. Inflamma Bowel Dis. 2008;14:147-161 10. Garrett WS, Lord G, Punit S, et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell. 2007;131:33-45 157 11. Sellon R, Tonkonogy S, Schultz M, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66:5224-5231 12. Sadlack B, Merz H, Schorle H et al. Ulcerative colitis-like disease in mice with a disrupted interleukin-2 gene. Cell. 1993;75:203-261 13. Kim SC, Tonkonogy SL, Karrasch T et al. Dual-association of gnotobiotic IL-10-/- mice with 2 nonpathogenic commensal bacteria induces aggressive pancolitis. Inflamm Bowel Dis. 2007;13:1457-1466 14. Warren S. Why study animal models of IBD? Inflamm Bowel Dis. 2008;14:S129-S131 15. Wirtz S, Neufert C, Weigmann B, et al. Chemically induced mouse model of intestinal inflammation. Nat Protoc. 2007;2:541-546 16. Keuhl CJ, Wood HD, Marsh TL, et al. Colonization of the Cecal Mucosa by Helicobacter hepaticus Impacts the Diversity of the Indigenous Microbiota. Infect Immun. 2005;73:6952-6961 17. Pratt JS, Sachen KL, Wood HD, et al. Modulation of host immune responses by the Cytolethal Distending Toxin of Helicobacter hepaticus. Infect Immun. 2006;74:4406-4503 18. Asquith MJ, Boulard O, Powrie F et al. Pathogenic and protective roles of MyD88 in leukocytes and epithelial cells in mouse models of inflammatory bowel disease. Gastroenterol. 2010;139:519-529 19. Dieleman L, Arends A, Tonkonogy S et al. Helicobacter hepaticus does not induce or potentiate colitis in interleukin-10-deficient mice. Infect Immun. 2000;68:5107-5113 20. Antonopoulos D, Huse S, Morrison H, et al. Reproducible community dynamics of the gastrointestinal microbiota following antibiotic perturbation. Infect immun. 2009;77:2367-2375 21. Robinson C,, Young VB. Antibiotic administration alters the community structure of the gastrointestinal microbiota. Gut Microbes. 2010;1:1-6 158 22. Young VB, Knox KA, Pratt JS, et al. In Vitro and In Vivo Characterization of Helicobacter hepaticus Cytolethal Distending Toxin Mutants. Infect Immun. 2004;72:2521-2527 23. Young VB, Schimdt TM. Antibiotic-Associated Diarrhea Accompanied by Large-Scale Alterations in the Composition of the Fecal Microbiota. J Clin Microbiol. 2004;42:1203-1206 24. Cole JR, Chai B, Farris RJ, et al. The ribosomal database project (RDP-II): introducing myRDP space and quality controlled public data. Nucl Acids Res. 2007;35:D169-172 25. Schloss P, Westcott S, Ryabin T, et al. Introducing mothur: Open Source, Platform-independent, Community-supported Software for Describing and Comparing Microbial Communities. Appl Environ Microbiol. 2009;23:7537-7541 26. Colwell RK. EstimateS: Statistical estimation of species richness and shared species from samples. User's Guide and application published at: http://purloclcorg/estimates. 2006 27. Kumar S, Tamura K, Nei M. MEGA3: integrated software for molecular evolutionary genetics analysis and sequence alignment. . Brief Bioinform 2004;5:150-163 28. Martin FE, Nadkarni MA, Jacques NA, et al. Quantitative Microbiological Study of Human Carious Dentine by Culture and Real-Time PCR: Association of Anaerobes with Histopathological Changes in Chronic Pulpitis. J Clin Microbiol. 2002;40:1698-1704 29. Nitsche A, Becker M, Junghahn I, et al. Quantification of human cells in NOD/SCID mice by duplex real-time polymerase-chain reaction. Haematologica. 2001;86:693-699 30. Lees CW, Zacharias WJ, Tremelling M, et al. Analysis of Germline GLI1 Variation Implicates Hedgehog Signalling in the Regulation of Intestinal Inflammatory Pathways. PLoS Med. 2008;5:1761-1775 31. Pfaffl MW. A new mathematical model for relative quantification in realtime RT-PCR. Nucleic Acids Res. 2001;29:e45 159 32. Guide to Performing Relative Quantitation of Gene Expression Using Real-Time Quantitative PCR. Applied Biosystems. 2004 33. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nature Protoc. 2008;3:1101-1108 34. White JR, Nagarajan N, Pop M. Statistical methods for detecting differentially abundant features in clinical metagenomic samples. PLoS Comput Biol. 2009;5:e1000352 35. Maslowski KM, Vieira AT, Ng A, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282-1286 36. Wostmann B. The germfree animal in nutritional studies. Ann Rev Nutri. 1981;1:257-279 37. Smith K, McCoy KD, Macpherson AJ. Use of axenic animals in studying the adaptation of mammals to their commensal intestinal microbiota. Semin Immunol. 2006;19:59-69 38. Round J, Mazmanian S. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol. 2009;9:313-323 39. Ott SJ, Musfeldt M, Wenderoth DF, et al. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004;53:685-693 40. Nishikawa J, Kudo T, Sakata S, et al. Diversity of mucosa-associated microbiota in active and inactive ulcerative colitis. Scandinavian J Gastroenterol. 2009;44:180-186 41. Manichanh C, Rigottier-Gois L, Bonnaud E, et al. Reduced diversity of faecal microbiota in Crohn's disease revealed by a metagenomic approach. Gut. 2006:205-211 42. Scanlan PD, Shanahan F, O'Mahony C, et al. Culture-Independent Analyses of Temporal Variation of the Dominant Fecal Microbiota and Targeted Bacterial Subgroups in Crohn's Disease. J Clin Microbiol. 2006;44:3980-3988 160 43. Rehman A, Lepage P, Nolte A, et al. Transcriptional activity of dominant gut mucosal microbiota in chronic inflammatory bowel diseases. J Med Microbiol. 2010;59:1114-1122 44. Takaishi H, Matsuki T, Nakazawa A, et al. Imbalance in intestinal microflora constitution could be involved in the pathogenesis of inflammatory bowel disease. Int J Med Microbiol. 2008;298:463-472 45. Muniyappa P, Gulati R, Mohr F, et al. Use and safety of rifaximin in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2009;49:400-404 46. Dickinson RJ, O'Connor HJ, Pinder I, et al. Double blind controlled trial of oral vancomycin as adjunctive treatment in acute exacerbations of idiopathic colitis. Gut. 1985;26:1380-1384 47. Nilsson H-O, Ouis I-S, Stenram U, et al. High Prevalence of Helicobacter Species Detected in Laboratory Mouse Strains by Multiplex PCR-Denaturing Gradient Gel Electrophoresis and Pyrosequencing. J Clin Microbiol. 2004;42:3781-3788 48. Li Z, Stephen JD, Martin G, et al. Natural Colonization with Helicobacter species and the Development of Inflammatory Bowel Disease in Interleukin-10deficient Mice. Helicobacter. 2005;10:223-230 49. Taylor NS, Xu S, Nambiar P, et al. Enterohepatic Helicobacter Species Are Prevalent in Mice from Commercial and Academic Institutions in Asia, Europe, and North America. J Clin Microbiol. 2007;45:2166-2172 50. Mazmanian S, Round J, Kasper D. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453:620-625 51. Cahill R, Foltz C, Fox J, et al. Inflammatory bowel disease: an immunitymediated condition triggered by bacterial infection with Helicobacter hepaticus. Infect Immun. 1997;65:3126-3131 52. Li X, Fox JG, Whary MT, et al. SCID/NCr Mice Naturally Infected with Helicobacter hepaticus Develop Progressive Hepatitis, Proliferative Typhlitis, and Colitis. Infect Immun. 1998;66:5477-5484 161 53. Chow J, Mazmanian S. A Pathobiont of the Microbiota Balances Host Colonization and Intestinal Inflammation. Cell host Microbe. 2010;7:265-276 54. Foltz CJ, Fox JG, Yan L, et al. Evaluation of various oral antimicrobial formulations for eradication of Helicobacter hepaticus. Lab Anim Sci. 1996;46:193-197 55. Madsen KL, Doyle JS, Tavernini MM, et al. Antibiotic therapy attenuates colitis in interleukin 10 gene-deficient mice. Gastroenterol. 2000;118:1094-1105 56. Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the Human Intestinal Microbial Flora. Science. 2005;308:1635-1638 57. Sekirov I, Tam NM, Jogova M, et al. Antibiotic-Induced Perturbations of the Intestinal Microbiota Alter Host Susceptibility to Enteric Infection. Infect Immun. 2008;76:4726-4736 58. De La Cochetiere MF, Durand T, Lepage P, et al. Resilience of the Dominant Human Fecal Microbiota upon Short-Course Antibiotic Challenge. J Clin Microbiol. 2005;43:5588-5592 59. Garner C, Antonopoulos D, Wagner B, et al. Perturbation of the small intestine microbial ecology by streptomycin alters pathology in a Salmonella enterica serovar typhimurium murine model of infection. Infect Immun. 2009;77:2691-2702 60. Stecher B, Macpherson AJ, Hapfelmeier S, et al. Comparison of Salmonella enterica Serovar Typhimurium Colitis in Germfree Mice and Mice Pretreated with Streptomycin. Infect Immun. 2005;73:3228-3241 61. Croswell A, Amir E, Teggatz P, et al. Prolonged impact of antibiotics on intestinal microbial ecology and susceptibility to enteric Salmonella infection. Infect Immun. 2009;77:2741-2753 62. Chin AC, Parkos CA. Neutrophil transepithelial migration and epithelial barrier function in IBD: potential targets for inhibiting neutrophil trafficking. Ann NY Acad Sci. 2006;1072:276-287 162 63. Spehlmann ME, Begun AZ, Burghardt J, et al. Epidemiology of inflammatory bowel disease in a German twin cohort: results of a nationwide study. Inflamm Bowel Dis. 2008;14:968-976 64. Borody TJ, Warren EF, Leis S, et al. Treatment of ulcerative colitis using fecal bacteriotherapy. J Clin Gastroenterol. 2003;37:42-47 65. Haskey N, Dahl WJ. Synbiotic therapy: a promising new adjunctive therapy for ulcerative colitis. Nutr Rev. 2006;64:132-138 66. Erdman SE, Rao VP, Poutahidis T, et al. Nitric oxide and TNF-alpha trigger colonic inflammation and carcinogenesis in Helicobacter hepaticusinfected, Rag2-deficient mice. PNAS. 2009;106:1027-1032 67. Brandl K, Plitas G, Mihu C, et al. Vancomycin-resistant enterococci exploit antibiotic-induced innate immune deficits. Nature. 2008;455:804-807 68. Ivanov II, Frutos RL, Manel N, et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell host Microbe. 2008;4:337-349 69. Kullberg MC, Rothfuchs AG, Jankovic D, et al. Helicobacter hepaticusInduced Colitis in Interleukin-10-Deficient Mice: Cytokine Requirements for the Induction and Maintenance of Intestinal Inflammation. Infect Immun. 2001;69:4232-4241 70. Mizoguchi E. Chitinase 3-like-1 exacerbates intestinal inflammation by enhancing bacterial adhesion and invasion in colonic epithelial cells. Gastroenterol. 2006;130:398-411 71. Wlodarska M, Finlay BB. Host immune response to antibiotic perturbation of the microbiota. Mucosal Immunol. 2010;3:100-103 163 Chapter 5 Summary, synthesis and future directions Understanding the changes that occur during onset and progression of disease can also allow for predictive diagnoses from biopsies or fecal samples and help in managing disease. Since it is difficult to investigate changes in IBD in patients due to ethical considerations and inability to predict onset of disease, we must rely on animal models. However, studies on changes in the microbiota associated with IBD in murine models are very sparse. The purpose of the work present here was to investigate the role of the microbiota at various stages of development of IBD through use of mouse models of disease. In order to investigate the extent to which the microbiota is involved in disease development, two models were used to address two different questions: First, DSS-induced colitis is commonly used to investigate onset of IBD, however, information of microbiota changes that may occur was limited (1, 2). Previously, there was one study, using non-culture based methods, that documented an altered microbiota during DSS-induced colitis (2), however, the role of the microbiota at early stages of disease were unknown. We therefore conducted a temporal study to document changes that occur during early and late stages of disease. The current study demonstrates that community changes are not only associated with later stages of disease, but these changes also appear very 164 early. Alterations in the microbial community occurred as soon as three days post DSS administration, just as host inflammatory responses were also observed. This altered structure persisted to 14 days post DSS administration as inflammatory changes progressed and became more severe. Microbial changes in community structure were associated with changes in host response, linking disease with shifts in microbial community structure at various stages of disease progression. These results indicate that microbiota changes are indeed associated with DSS-induced colitis, and these changes may also be associated with maintenance of inflammation. In the second model, it is known that H. hepaticus alters the microbiota as it becomes a predominant member of the microbial community (3), thus, the model inherently involves changes in the microbiota that triggers disease. However, we did not know whether changes in the resident community would exacerbate or reduce disease once initiated, therefore, the role of the microbiota in this system was further probed by finding by inducing shifts in the resident community by antibiotic treatments. We found that shifts in the resident microbiota, or changes in prevalence of H. hepaticus in the community, did not influence severity of disease. We thus demonstrated that the role of the resident microbiota might be minor once disease is initiated in this model. Also, we investigated whether the microbiota was important in disease induction in mice in our colonies through infection of germfree mice with H. hepaticus. Results revealed that the cecum sections in mice monoassociated with H. hepaticus were similar to uninfected controls indicating that the microbiota was essential in initiating disease. Here, it 165 appeared that the microbiota is essential for initiating disease but not in exacerbating or reducing inflammation during disease progression. It is thus apparent that there are different roles for the microbiota in different stages of IBD, and these roles may be dependent on other factors associated with IBD such as environmental triggers as in the case of H. hepaticus infection. Elucidating these roles of the microbiota associated to IBD will be important in developing methods for prognosis and/or treatment of human disease. Future Directions From our work and that of others, we know that the microbiota can be essential in initiation of disease as seen in H. hepaticus triggered disease in IL-10 deficient mice, but play a lesser role in exacerbation/reduction of disease as demonstrated here. We also show alterations in the microbiota can play a role at the onset and persistence of disease, as shown by microbiota changes associated with DSSinduced colitis. However, even with these two models, several aspects by which the microbiota is involved in disease development are yet to be determined (table 5.1). The following attempts to present studies that may fill these gaps in knowledge. 166 Stages of IBD Initiation DSS-induced colitis Maybe [(4-6)] H. hepaticus triggered IBD Yes [Chapter 4, (7)] Progression Maybe [Chapter 3] Unknown Exacerbation/ Reduction Yes [(8)] No [Chapter 4] Table 5.1 Roles of the microbiota in at various stages of inflammatory bowel disease. 167 The role of the microbiota in initiation of IBD In H. hepaticus triggered IBD, there is a clear role for the microbiota in initiation of disease since germfree mice monoassociated with this bacterium do not develop inflammation. However, the role microbes play in DSS-induced colitis is unclear. In some studies, DSS administration resulted in increased inflammation in germfree mice (5), indicating a protective function of the microbiota in onset of disease, while others show more severe disease phenotype in conventional animals compared to germfree counterparts (4), indicating a detrimental role of the microbiota leading to onset of disease. Nonetheless, differences in disease development in germfree and conventional animals demonstrate a role of the microbiota in affecting DSS-induced disease. However, due to conflicting results, more studies are necessary in order to understand the role of the microbiota in this model. Some of this conflicting results in germfree mice investigations stem from the use of different strains of mice (9), ages and even diet. To investigate the role of the microbiota in disease initiation, several strains of germfree mice, at similar ages, can be given DSS and then monitored for disease development. It will be important to provide elemental diets (minerals, vitamins, amino acids, fats and sugars), which are devoid of potential antigenic material such as bacterial components, plant material and other proteins. If inflammation does not develop in these germfree mice, then they can be conventionalized before, after and during DSS administration to observe effects of introduced microbiota on disease development. If disease manifestations were altered in any way after introduction 168 of the microbiota, this would indicate that microbes play a role in initiating disease. Similar experimental designs have been used in testing the effects of probiotics and antibiotics in IBD (8, 10). Alternatively, inflammation may be worse in germfree mice given DSS suggesting that the microbiota has a protective role against disease induction. Testing whether live bacteria or heat-killed bacteria impart these benefits can then be done. In conventionally raised mice, DSS-induced disease is known to be highly strain dependent (9, 11, 12). This may be related to the microbiota harbored by various strains since it is known that the microbial community differs according to many factors, including the host (13, 14). Therefore, characterizing the microbiota of various strains that are associated with different disease manifestations may reveal particular community structures linked to disease. If a particular microbial community is associated with severe inflammation, transfer of this microbiota into another strain can be done to test if the community is truly involved in IBD in this model. Others have performed similar experiments in the T-bet Rag-/- ulcerative colitis (TRUC) model where the microbiota of inflamed mice were transmitted to wild type mice that subsequently developed disease (15), indicating that genetic background can alter the microbiota to a community that can induce IBD. The role of the microbiota in disease progression of IBD There has been some evidence that the microbiota in DSS-induced colitis is important in sustaining inflammation since antibiotics have been successful in 169 ameliorating disease (8). However, the extent to which microbes are important in disease in H. hepaticus triggered infection is unknown. In H. hepaticus triggered disease, we know that H. hepaticus is important in maintaining inflammation since eradication of H. hepaticus ameliorates disease. Here, we also showed that changes in the community do not affect disease manifestation. However, whether the resident community is at all necessary, after disease is already induced, is unknown. Cefoperazone (0.5 mg/ml) can provide a means to test whether the microbiota is important in potentiating disease since it significantly reduces the intestinal bacteria more than 1000 fold (16). Unfortunately, this antibiotic concentration also eradicates H. hepaticus (see appendices 1 and 2). To circumvent this issue, heat-killed Helicobacter may be useful. Stimulation from Helicobacter is necessary in this disease model, but whether live bacteria are needed has yet to be determined. By administering heat-killed Helicobacter to conventional mice can address this question. Subsequently, if disease is initiated using the heat-killed bacteria, cefoperazone can be administered to mice in conjunction with killed bacteria to investigate effects of significant reduction in microbiota during disease progression. With this experimental design, the resident microbiota can be significantly reduced while maintaining stimulation by H. hepaticus. Amelioration of disease by cefoperazone treatment would indicate that the microbiota is essential in reducing or maintaining disease in this model. If, however, disease progresses unaltered during cefoperazone treatment, it may be because the microbiota is not essential for 170 disease perpetuation once it has been induced. Testing whether the microbiota is essential in disease perpetuation would be challenging since removing the microbiota from a conventional animal is impossible. However, by performing gnotobiotic experiments and finding out whether a simple community is sufficient to trigger disease in this model, other experiments may be possible (see upcoming section). For instance, if disease can be initiated in gnotobiotic communities consisting of bacteria that are transient in the intestinal community such as those present in probiotic mixtures (17), we can investigate whether H. hepaticus triggered disease perpetuates once the transient microbes have been cleared. Testing the importance of microbiota complexity in IBD It is not known whether a complex microbial community is necessary to induce disease, or whether a simple community is sufficient. Murine association with the Altered Schaedler Flora (ASF) can be used as a simple community to investigate changes in the microbiota that may accompany disease in DSS-induced colitis and H. hepaticus triggered disease in IL-10 deficient mice. Development of disease in mice harboring simple communities would indicate that complex communities are unnecessary; a single microbe may even be enough for inflammation to occur. Bacteroides thetaiotaomicron is part of the normal human microbiota (18), and thought to stimulate intestinal development in germfree mice so it resembles that 171 of conventional animals (6). Due to its non-pathogenic nature, as well as its benefits in attenuating some characteristics of germfree animals, including vascularization, architecture and immune modulation (6, 19), this bacterium can be used to infect germfree mice before DSS treatment, or H. hepaticus infection, to test whether a simple system of bacterial-host interaction is enough to trigger disease comparable to that seen in conventionally raised mice. If these monoassociated mice develop disease that is similar to that observed in their conventionalized counterparts in each IBD model system, this would imply that simple microbial interaction or even luminal antigenic stimulation from killed bacteria, or bacterial components, might be enough to cause inflammation in the DSS-induced model, as observed in other systems (20). The next step would involve inoculation of mice with heat-killed bacteria and observing whether disease occurs in these models. DSS-induced colitis: Do alterations in the microbiota precede immune changes? To attempt to tease apart changes induced by DSS administration, a study can be conducted where frequent samples are collected between day zero and day three of DSS administration. Recent evidence indicates that intimate interactions between the microbiota and host occur within the first 24 hours of DSS administration (21). Bacteria were present in the normally impenetrable mucosa layer indicating DSS increases permeability of the mucin matrix allowing close association with the host within a day, before host inflammatory responses were 172 observed (21). By investigating whether changes in the microbial community structure occurred within this time would be helpful in tracing the sequence of events involving host response and microbiota changes after exposure to DSS. If changes in host response occur before alteration in the microbiota, it is likely that DSS acts directly on the host in the presence of luminal bacteria and community shifts are not involved in onset of disease. Subsequent changes in the microbial community may simply be a reaction to host’s inflammatory response (22), as it known that components of the immune system can alter the microbiota. This is exemplified by increases in segmented filamentous bacteria in mice with deficient expression of intestinal IgA (23). Subsequent reconstitution of IgA in intestinal tissue resulted in a return to a community resembling that in mice expressing IgA (23). Alternatively, if changes in the microbial community structure occur before changes in the host immune system are detected, this may be an indication that the altered microbiota plays a role in disease causation. Effects of different microbial structures on development of disease were tested by antibiotic treatment (8), but using gnotobiotic models would also be useful in understanding the role of certain members of the community, as discussed above. However, due to the almost simultaneous changes in the microbiota and host immune response, teasing apart the sequence of event may be challenging. Germfree and gnotobiotic experiments proposed at the beginning of this section 173 may be more prudent approaches to investigating the role of the microbiota in onset of disease. Conclusion Over the past two decades, studies on the microbiota and its influence on disease development has grown exponentially (24). Due to the complexities involved in understanding both the role of the hosts’ immune response and changes in diversity and function of the microbiota, studies that lay ahead will be challenging. Nonetheless, development of new tools for investigation of these changes, and continued vigorous research into the field, provide a promising future for patients affected with diseases such as IBD. 174 REFERENCES 175 REFERENCES 1. Araki Y, Andoh A, Tsujikawa T, et al. Alterations in intestinal microflora, faecal bile acids and short chain fatty acids in dextran sulphate sodium-induced experimental acute colitis in rats. Eur J Gastroenterol Hepatol. 2001;13:107-112 2. Heimesaat M, Fischer A, Siegmund B, et al. Shift towards proinflammatory intestinal bacteria aggravates acute murine colitis via Toll-like receptors 2 and 4. PLoS ONE. 2007;25:e662 3. Keuhl CJ, Wood HD, Marsh TL, et al. Colonization of the Cecal Mucosa by Helicobacter hepaticus Impacts the Diversity of the Indigenous Microbiota. Infect Immun. 2005;73:6952-6961 4. Hudcovic T, Stepankova R, Cebra J, et al. The Role of Microflora in the Development of Intestinal Inflammation: Acute and Chronic Colitis Induced by Dextran Sulfate in Germ-Free and Conventionally Reared Immunocompetent and Immunodeficient Mice. Folia Microbiol. 2001;46:565-572 5. Kitajima S, Morimoto M, Sagara E, et al. Dextran Sodium Sufate-Induced Colitis in Germ-Free IQI/Jic Mice. Exp Anim. 2001;50:387-395 6. Stappenbeck T, Hooper L, Gordon J. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. PNAS. 2002;99:15451-15455 7. Dieleman L, Arends A, Tonkonogy S, et al. Helicobacter hepaticus does not induce or potentiate colitis in interleukin-10-deficient mice. Infect Immun. 2000;68:5107-5113 8. Rath H, Schultz M, Freitag R, et al. Different Subsets of Enteric Bacteria Induce and Perpetuate Experimental Colitis in Rats and MIce. Infect Immun. 2001;69:2277-2285 9. Mahler M, Bristol JI, Leiter HE, et al. Differential susceptibility of inbred mouse strains to dextran sulphate sodium-induced colitis. Am J Physiol. 1998;274:544-551 176 10. Herias MV, Koninkx JF, Vos JG, et al. Probiotic effects of Lactobacillus casei on DSS-induced ulcerative colitis in mice. Int J Food Microbiol. 2005;103:143-155 11. Suzuki R, Kohno H, Sugie S, et al. Strain differences in the susceptibility to azoxymethane and dextran sodium sulfate-induced colon carcinogenesis in mice. Carcinogenesis. 2006;27:162-169 12. Stevceva L, Pavli P, Buffinton G, et al. Dextran sodium sulphate-induced colitis activity varies with mouse strain but develops in lipopolysaccharideunresponsive mice. J Gastroentorol Hepatol. 1999;14:54-60 13. Rawls JF, Samuel BS, Gordon JI. Gnotobiotic zebrafish reveal evolutionarily conserved responses to the gut microbiota. PNAS. 2004;101:45964601 14. Ley RE, Peterson DA, Gordon JI. Ecological and Evolutionary Forces Shaping Microbial Diversity in the Human Intestine. Cell. 2006;24:837-848 15. Garrett WS, Lord G, Punit S, et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell. 2007;131:33-45 16. Antonopoulos D, Huse S, Morrison H, et al. Reproducible community dynamics of the gastrointestinal microbiota following antibiotic perturbation. Infect Immun. 2009;77:2367-2375 17. Gaudier E, Michel C, Segain J-P, et al. The VSL#3 Probiotic Mixture Modifies Microflora but Does Not Heal Chronic Dextran-Sodium Sulfate-Induces Colitis or Reinforce the Mucus barrier in Mice. J Nutr. 2005;135:2753-2761 18. Duerden BI. The isolation and identification of Bacteroides spp. from the normal human faecal flora. J Med Microbiol. 1980;13:69-78 19. Peterson DA, McNulty NP, Guruge JL, et al. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe. 2007;2:328-339 20. Hoffmann M, Kim SC, Sartor RB, et al. Enterococcus faecalis strains differentially regulate Alix/AIP1 protein expression and ERK 1/2 activation in 177 intestinal epithelial cells in the context of chronic experimental colitis. J Proteome Res. 2009;8:1183-1192 21. Png CW, Linden SK, Gilshenan KS, et al. Mucolytic Bacteria With Increased Prevalence in IBD Mucosa Augment In Vitro Utilization of Mucin by Other Bacteria. Am J Gastroenterol. 2010;e-pub 22. Salzman NH, Underwood MA, Bevins CL. Paneth cells, defensins, and the commensal microbiota: a hypothesis on intimate interplay at the intestinal mucosa. Semin Immunol. 2007;2007:2 23. Suzuki K, Meek B, Doi Y, et al. Aberrant expansion of segmented filamentous bacteria in IgA-deficient gut. PNAS. 2004;101:1981-1986 24. Sekirov I, Russell SL, Antunes LC, et al. Gut Microbiota in Health and Disease. Physiol Rev.90:859-904 178 APPENDICES 179 Appendix A The effects of cefoperazone on Helicobacter hepaticus colonization. Purpose Cefoperazone (0.5 mg/ml) was previously used to alter microbial community structure in the intestine (1), and is commonly used in selective media to screen for H. hepaticus growth in vitro (2). We therefore initially used this antibiotic to alter the microbial community in mice infected with H. hepaticus in order to study effects of changes in the community during disease. Surprisingly however, cefoperazone treatment eradicated H. hepaticus in vivo. After reducing the concentration administered to mice by ten fold, there seemed to be negligible effects on total bacterial and H. hepaticus loads in vivo. We therefore suspected that bacterial colonization maybe dependent on concentration of cefoperazone administered. We thus tested this hypothesis that changes in the microbiota are associated with cefoperazone concentration administered. Here, effects of various concentrations of cefoperazone on total bacterial load and H. hepaticus loads are reported. Methods and materials To investigate the effects of cefoperazone treatment on H. hepaticus in vivo eleven groups of mice were infected with H. hepaticus for ten days before 180 antibiotic was administered. There were three mice in each group. Dosage ranged from 0.5 mg/ml to 0.05 mg/ml. Previously, 0.05 mg/ml was seen to have no effect of either H. hepaticus colonization or total bacterial load. Mice were sacrificed seven days after the start of cefoperazone administration as shown in figure A1.1. Quantitative PCR was performed and clone libraries constructed from mice from each experimental group as described in previous chapters 3 and 4. 181 C57BL/6 mice gavage with H. hepaticus 0 mg/ml 0.05 mg/ml 0 mg/ml 0.20 mg/ml 10 days 0.30 mg/ml 0.40 mg/ml 0.50 mg/ml 0.10 mg/ml 0.25 mg/ml 0.35 mg/ml 0.45 mg/ml 7 days Harvest cecum for microbial ecology analysis Figure A1.1 Experimental design depicting administration of various cefoperazone concentrations to H. hepaticus infected mice. 182 Results and conclusions Quantitative data shows that 0.5 mg/ml to 0.30 mg/ml significantly reduced total bacterial loads over 1000 fold and eradicated H. hepaticus. At about 0.25 mg/ml, the antibiotic did not affect bacterial load or presence of H. hepaticus, indicating a threshold for cefoperazone activity against intestinal bacteria. Eradication of H. hepaticus was confirmed by allowing the intestinal communities to recover after cefoperazone administration. It was found that H. hepaticus was not detectable after four weeks cessation of antibiotic administration. 183 Fold change compared to no cefoperazone treatment 1000 0.50 0.45 0.40 0.35 0.30 0.25 0.20 0.10 0.05 0 cefoperazone (mg/ml) -1000 H. hepaticus Total bacteria -2000 -3000 -4000 -5000 -6000 -7000 Figure A1.2 Cefoperazone concentrations above 0.25 mg/ml cause significant decreases in total bacterial loads and eradicates H. hepaticus. 184 To investigate these affects more closely, clone libraries were constructed. Clone libraries also show presence of H. hepaticus in communities in mice treated with up to 0.25 mg/ml cefoperazone. The community in mice treated with 0.05 mg/ml cefoperazone appeared to be more similar to communities in untreated mice (figure A1.2 A). Furthermore, although bacterial load in mice treated with 0.05 mg/ml was comparable to that in mice treated with 0.10 mg/ml cefoperazone, there was a decrease in richness in the latter community (figure A1.2 B), indicating that this concentration affects colonization of some members of the community more than others. With 0.30 mg/ml and more, decrease in bacterial loads made it very difficult to construct clone libraries due to poor primary amplification of the bacterial 16S rRNA gene. This indicates that these higher concentrations affect many members of the intestinal community than 0.25 mg/ml cefoperazone. Perhaps the machinery designed to remove cefoperazone from the bacterial cells are overwhelmed at these concentrations causing the bacteria to be susceptible to the antibiotic. 185 100% 90% Clostridiaceae unclassified_Peptococcaceae Peptococcaceae Incertae Sedis XIII Erysipelotrichaceae Helicobacteraceae unclassified_Porphyromonadaceae Porphyromonadaceae unclassified_"Ruminococcaceae" Ruminococcaceae Anaeroplasmataceae unclassified_"Lachnospiraceae" Lachnospiraceae 80% Prevalence of bacteria 70% 60% 50% 40% 30% 20% 10% 0% 0 0 uninfected 0.05 0.1 0.2 0.25 cefoperazone (mg/ml) 60 0 mg/ml cef, no H. hepaticus 0 mg/ml cef, H. hepaticus 0.20 mg/ml cef, H. hepaticus 0.25 mg/ml cef, H. hepaticus 0.05 mg/ml cef, H. hepaticus 0.10 mg/ml cef, H. hepaticus 50 OTU 40 30 20 10 0 0 20 40 60 80 100 Observed sequences Figure A1.3. (A) H. hepaticus were present in the microbiota of communities treated with cefoperazone concentrations below 0.25 mg/ml. Some communities showed significant changes due to H. hepaticus infection and cefoperazone treatment. (B) There was also a reduction in intestinal community richness when concentrations of 0.20 mg/ml cefoperazone and above were administered to mice. For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. 186 At 0.03 mg/ml cefoperazone, H. hepaticus is absent from microbial communities. Below 0.25 mg/ml, however, H. hepaticus is present, and together with the finding that total bacterial load is also unaffected at this concentration, it can be postulated that cefoperazone is not active at lower concentrations. This may be due to break down of the compound at low concentrations during in vivo passage since typical signs of cefoperazone treatment, such as dark cecal contents and distended cecum, were absent from these mice (see photographs in appendix B). To test this, cefoperazone levels can be measures from the cecal contents by mass spectrometry. Another possibility is that at 0.30 mg/ml, H. hepaticus is eradicated from the intestinal community suggesting that the bacterium is susceptible to this concentration of cefoperazone. H. hepaticus is known to have efflux pumps that functions to remove antibiotics from the bacterial cell (3). It is easy to imagine that by increasing cefoperazone levels, that the pump may not efficiently remove the molecules before they act. To test the hypothesis that an efflux pump may be responsible for susceptibility of H. hepaticus to high concentrations of cefoperazone, mutants with increased pumping activity can be used to test whether they are resistant to higher concentrations of cefoperazone treatment. Alternatively, efflux pump inhibitors may be used, in a dose-dependent manner, to investigate whether there are effects on the ability of H. hepaticus to grow at concentrations below 0.25 mg/ml (4). If pumps are indeed important in conferring resistance to cefoperazone in this bacterium, increasing levels of the inhibitor increase susceptibility. 187 Dosages of 0.10 mg/ml to 0.25 mg/ml were associated with higher relative abundance of H. hepaticus. More experiments to investigate how H. hepaticus colonizes a community is needed to understand the low relative abundance of the bacterium in communities associated with 0.05 mg/ml cefoperazone administration compared to those administered 0.10, 0.20 and 0.25 mg/ml cefoperazone. It would be expected that H. hepaticus would be more prevalent at lower antibiotic concentrations, with communities resembling those of either communities from mice not administered cefoperazone; or more like those associated with 0.10 mg/ml. Perhaps at 0.05 mg/ml cefoperazone treatment, resident microbes shifts so certain members of particular families increase in abundance. This may explain lowered relative abundances of H. hepaticus while total loads are unchanged, as well as richness (Shannon index=3.27) of the microbiota similar to that in cefoperazone untreated mice (Shannon index= 3.37), compared to the low richness of antibiotic treated mice (Shannon index= 1.59 with 0.10 mg/ml cefoperazone, 1.52 with 0.20 mg/ml cefoperazone and 1.51 with 0.25 mg/ml cefoperazone) at 3% cutoff. 188 REFERENCES 189 REFERENCES 1. Antonopoulos D, Huse S, Morrison H, et al. Reproducible community dynamics of the gastrointestinal microbiota following antibiotic perturbation. Infect Immun. 2009;77:2367-2375 2. Pratt JS, Sachen KL, Wood HD, et al. Modulation of host immune responses by the Cytolethal Distending Toxin of Helicobacter hepaticus. Infect Immun. 2006;74:4406-4503 3. Belzer C, Stoof J, Breijer S, et al. The Helicobacter hepaticus hefA gene is involved in resistance to amoxicillin. Helicobacter. 2009;14:72-79 4. Lomovskaya O, Warren MS, Lee A, et al. Identification and Characterization of Inhibitors of Multidrug Resistance Efflux Pumps in Pseudomonas aeruginosa: Novel Agents for Combination Therapy. Antimicrob Agents Chemother. 2001;45:105-116 190 Appendix B The effects of cefoperazone administration in H. hepaticus triggered disease in IL-10 defiecient mice. Purpose Others have shown effects of antibiotic treatment on H. hepaticus infected mice and reported that some are effective in ridding colonies of this bacterium (1). We showed that cefoperazone, an antibiotic normally administered through intravenously in humans, was found to eradicate H. hepaticus in wild type mice (appendix A). However, effect of cefoperazone treatment in H. hepaticus triggered disease was unknown. We therefore tested the hypothesis that cefoperazone administration was attenuate disease in IL-10 deficient mice infected with H. hepaticus. Methods and materials Four groups of mice were used in this experiment as indicated in figure A2.1. After euthanasia, the ceca of the mice were removed and used for genomic DNA extraction and histologic preparation (see chapters 3 and 4 of this thesis). 191 Figure A2.1 Experimental design showing H. hepaticus infection prior to cefoperazone treatment. 192 Results and Discussion Photographs were taken to document changes that occur with different treatments (figure A2.2). Cefoperazone treatment resulted in distension of the cecum with dark, fluid luminal content, making tissue fragile. This indicates that cefoperazone is associated with alterations in the host, which may be related to changes in the microbiota. Changes in host response mediators were observed after cefoperazone treatment and documented in experiments described in chapter 4 of this thesis. Some of these changes in during immune regulation may affect permeability of the epithelial wall, which may be associated fluid luminal content and distension (3). H. hepaticus infection was associated with thickening of the cecal tissue making it more rigid, with a reduction in luminal content. This is a common sign of disease as scarring occurs during IBD and was also observed in experiments described in chapter 4. Cefoperazone administration, following H. hepaticus infection, appeared similar to tissue from cefoperzone treated, H. hepaticus uninfected mice. This may be due to effects of cefoperazone-induced alterations in microbiota. Whether cefoperazone alone can induce host immune changes is yet to be determined. This can be investigated by administering cefoperazone to germfree mice. 193 Figure A2.2 Photographs of ceca removed from mice from four treatment groups. (Top left) Cecum is normal with luminal content of paste-like consistency, unlike cecum from mice infected with H. hepaticus that have thick tissue (top right). With cefoperazone administration (bottom panels), the cecum become distended and tissue fragile, filled with dark luminal fluid. For interpretation of the references to color in this and all other figures, the reader is referred to the electronic version of this dissertation. 194 After cefoperazone administration, H. hepaticus infected mice appeared to have less inflammation. This corresponded to a reduction in total bacterial load and eradication of H. hepaticus (figure A1.3). This finding is supported by reports that eradication of H. hepaticus can ameliorate disease in rodents (1). More animals would have to be used in this experiment to attain more statistical power to dram convincing conclusions. 195 Figure A2.3 show significant inflammation in mice infected with H. hepaticus. After administration of cefoperazone, inflammation was attenuated. 196 REFERENCES 197 REFERENCES 1. Foltz CJ, Fox JG, Yan L, et al. Evaluation of various oral antimicrobial formulations for eradication of Helicobacter hepaticus. Lab Anim Sci. 1996;46:193-197 2. Abdo Z, Schuette UME, Bent SJ, et al. Statistical methods for characterizing diversity of microbial communities by analysis of terminal restriction fragment length polymorphisms of 16S rRNA genes. Environ Microbiol. 2006;8:929-938 3. Ménard S, Cerf-Bensussan N, Heyman M. Multiple facets of intestinal permeability and epithelial handling of dietary antigens. Mucosal Immunol. 2010;3:247-259 198