@phdthesis{Ganesh2013, author = {Ganesh, Bhanu Priya}, title = {Host-microbe interactions in the inflamed gut}, url = {http://nbn-resolving.de/urn:nbn:de:kobv:517-opus-69558}, school = {Universit{\"a}t Potsdam}, year = {2013}, abstract = {Initiation and perpetuation of inflammatory bowel diseases (IBD) may result from an exaggerated mucosal immune response to the luminal microbiota in a susceptible host. We proposed that this may be caused either 1) by an abnormal microbial composition or 2) by weakening of the protective mucus layer due to excessive mucus degradation, which may lead to an easy access of luminal antigens to the host mucosa triggering inflammation. We tested whether the probiotic Enterococcus faecium NCIMB 10415 (NCIMB) is capable of reducing chronic gut inflammation by changing the existing gut microbiota composition and aimed to identify mechanisms that are involved in possible beneficial effects of the probiotic. To identify health-promoting mechanisms of the strain, we used interleukin (IL)-10 deficient mice that spontaneously develop gut inflammation and fed these mice a diet containing NCIMB (106 cells g-1) for 3, 8 and 24 weeks, respectively. Control mice were fed an identically composed diet but without the probiotic strain. No clear-cut differences between the animals were observed in pro-inflammatory cytokine gene expression and in intestinal microbiota composition after probiotic supplementation. However, we observed a low abundance of the mucin-degrading bacterium Akkermansia muciniphila in the mice that were fed NCIMB for 8 weeks. These low cell numbers were associated with significantly lower interferon gamma (IFN-γ) and IFN-γ-inducible protein (IP-10) mRNA levels as compared to the NCIMB-treated mice that were killed after 3 and 24 weeks of intervention. In conclusion, NCIMB was not capable of reducing gut inflammation in the IL-10-/- mouse model. To further identify the exact role of A. muciniphila and uncover a possible interaction between this bacterium, NCIMB and the host in relation to inflammation, we performed in vitro studies using HT-29 colon cancer cells. The HT-29 cells were treated with bacterial conditioned media obtained by growing either A. muciniphila (AM-CM) or NCIMB (NCIMB-CM) or both together (COMB-CM) in Dulbecco's Modified Eagle Medium (DMEM) for 2 h at 37 °C followed by bacterial cell removal. HT-29 cells treated with COMB-CM displayed reduced cell viability after 18 h (p<0.01) and no viable cells were detected after 24 h of treatment, in contrast to the other groups or heated COMB-CM. Detection of activated caspase-3 in COMB-CM treated groups indicated that death of the HT-29 cells was brought about by apoptosis. It was concluded that either NCIMB or A. muciniphila produce a soluble and heat-sensitive factor during their concomitant presence that influences cell viability in an in vitro system. We currently hypothesize that this factor is a protein, which has not yet been identified. Based on the potential effect of A. muciniphila on inflammation (in vivo) and cell-viability (in vitro) in the presence of NCIMB, we investigated how the presence of A. muciniphila affects the severity of an intestinal Salmonella enterica Typhimurium (STm)-induced gut inflammation using gnotobiotic C3H mice with a background microbiota of eight bacterial species (SIHUMI, referred to as simplified human intestinal microbiota). Presence of A. muciniphila in STm-infected SIHUMI (SIHUMI-AS) mice caused significantly increased histopathology scores and elevated mRNA levels of IFN-γ, IP-10, tumor necrosis factor alpha (TNF-α), IL-12, IL-17 and IL-6 in cecal and colonic tissue. The number of mucin filled goblet cells was 2- to 3- fold lower in cecal tissue of SIHUMI-AS mice compared to SIHUMI mice associated with STm (SIHUMI-S) or A. muciniphila (SIHUMI-A) or SIHUMI mice. Reduced goblet cell numbers significantly correlated with increased IFN-γ (r2 = -0.86, ***P<0.001) in all infected mice. In addition, loss of cecal mucin sulphation was observed in SIHUMI-AS mice. Concomitant presence of A. muciniphila and STm resulted in a drastic change in microbiota composition of the SIHUMI consortium. The proportion of Bacteroides thetaiotaomicron in SIHUMI, SIHUMI-A and SIHUMI-S mice made up to 80-90\% but was completely taken over by STm in SIHUMI-AS mice contributing 94\% to total bacteria. These results suggest that A. muciniphila exacerbates STm-induced intestinal inflammation by its ability to disturb host mucus homeostasis. In conclusion, abnormal microbiota composition together with excessive mucus degradation contributes to severe intestinal inflammation in a susceptible host.}, language = {en} } @phdthesis{Slezak2013, author = {Slezak, Kathleen}, title = {Impact of intestinal bacteria on the anatomy and physiology of the intestinal tract in the PRM/Alf mouse model}, url = {http://nbn-resolving.de/urn:nbn:de:kobv:517-opus-68946}, school = {Universit{\"a}t Potsdam}, year = {2013}, abstract = {Introduction: Intestinal bacteria influence gut morphology by affecting epithelial cell proliferation, development of the lamina propria, villus length and crypt depth [1]. Gut microbiota-derived factors have been proposed to also play a role in the development of a 30 \% longer intestine, that is characteristic of PRM/Alf mice compared to other mouse strains [2, 3]. Polyamines and SCFAs produced by gut bacteria are important growth factors, which possibly influence mucosal morphology, in particular villus length and crypt depth and play a role in gut lengthening in the PRM/Alf mouse. However, experimental evidence is lacking. Aim: The objective of this work was to clarify the role of bacterially-produced polyamines on crypt depth, mucosa thickness and epithelial cell proliferation. For this purpose, C3H mice associated with a simplified human microbiota (SIHUMI) were compared with mice colonized with SIHUMI complemented by the polyamine-producing Fusobacterium varium (SIHUMI + Fv). In addition, the microbial impact on gut lengthening in PRM/Alf mice was characterized and the contribution of SCFAs and polyamines to this phenotype was examined. Results: SIHUMI + Fv mice exhibited an up to 1.7 fold higher intestinal polyamine concentration compared to SIHUMI mice, which was mainly due to increased putrescine concentrations. However, no differences were observed in crypt depth, mucosa thickness and epithelial proliferation. In PRM/Alf mice, the intestine of conventional mice was 8.5 \% longer compared to germfree mice. In contrast, intestinal lengths of C3H mice were similar, independent of the colonization status. The comparison of PRM/Alf and C3H mice, both associated with SIHUMI + Fv, demonstrated that PRM/Alf mice had a 35.9 \% longer intestine than C3H mice. However, intestinal SCFA and polyamine concentrations of PRM/Alf mice were similar or even lower, except N acetylcadaverine, which was 3.1-fold higher in PRM/Alf mice. When germfree PRM/Alf mice were associated with a complex PRM/Alf microbiota, the intestine was one quarter longer compared to PRM/Alf mice colonized with a C3H microbiota. This gut elongation correlated with levels of the polyamine N acetylspermine. Conclusion: The intestinal microbiota is able to influence intestinal length dependent on microbial composition and on the mouse genotype. Although SCFAs do not contribute to gut elongation, an influence of the polyamines N acetylcadaverine and N acetylspermine is conceivable. In addition, the study clearly demonstrated that bacterial putrescine does not influence gut morphology in C3H mice.}, language = {en} } @phdthesis{Schumann2013, author = {Schumann, Sara}, title = {Influence of intestinal inflammation on bacterial protein expression in monoassociated mice}, url = {http://nbn-resolving.de/urn:nbn:de:kobv:517-opus-67757}, school = {Universit{\"a}t Potsdam}, year = {2013}, abstract = {Background: Increased numbers of intestinal E. coli are observed in inflammatory bowel disease, but the reasons for this proliferation and it exact role in intestinal inflammation are unknown. Aim of this PhD-project was to identify E. coli proteins involved in E. coli's adaptation to the inflammatory conditions in the gut and to investigate whether these factors affect the host. Furthermore, the molecular basis for strain-specific differences between probiotic and harmful E. coli in their response to intestinal inflammation was investigated. Methods: Using mice monoassociated either with the adherent-invasive E. coli (AIEC) strain UNC or the probiotic E. coli Nissle, two different mouse models of intestinal inflammation were analysed: On the one hand, severe inflammation was induced by treating mice with 3.5\% dextran sodium sulphate (DSS). On the other hand, a very mild intestinal inflammation was generated by associating interleukin 10-deficient (IL-10-/-) mice with E. coli. Differentially expressed proteins in the E. coli strains collected from caecal contents of these mice were identified by two-dimensional fluorescence difference gel electrophoresis. Results DSS-experiment: All DSS-treated mice revealed signs of a moderate caecal and a severe colonic inflammation. However, mice monoassociated with E. coli Nissle were less affected. In both E. coli strains, acute inflammation led to a downregulation of pathways involved in carbohydrate breakdown and energy generation. Accordingly, DSS-treated mice had lower caecal concentrations of bacterial fermentation products than the control mice. Differentially expressed proteins also included the Fe-S cluster repair protein NfuA, the tryptophanase TnaA, and the uncharacterised protein YggE. NfuA was upregulated nearly 3-fold in both E. coli strains after DSS administration. Reactive oxygen species produced during intestinal inflammation damage Fe-S clusters and thereby lead to an inactivation of Fe-S proteins. In vitro data indicated that the repair of Fe-S proteins by NfuA is a central mechanism in E. coli to survive oxidative stress. Expression of YggE, which has been reported to reduce the intracellular level of reactive oxygen species, was 4- to 8-fold higher in E. coli Nissle than in E. coli UNC under control and inflammatory conditions. In vitro growth experiments confirmed these results, indicating that E. coli Nissle is better equipped to cope with oxidative stress than E. coli UNC. Additionally, E. coli Nissle isolated from DSS-treated and control mice had TnaA levels 4- to 7-fold higher than E. coli UNC. In turn, caecal indole concentrations resulting from cleavage of tryptophan by TnaA were higher in E. coli Nissle- associated control mice than in the respective mice associated with E. coli UNC. Because of its anti-inflammatory effect, indole is hypothesised to be involved in the extension of the remission phase in ulcerative colitis described for E. coli Nissle. Results IL-10-/--experiment: Only IL-10-/- mice monoassociated with E. coli UNC for 8 weeks exhibited signs of a very mild caecal inflammation. In agreement with this weak inflammation, the variations in the bacterial proteome were small. Similar to the DSS-experiment, proteins downregulated by inflammation belong mainly to the central energy metabolism. In contrast to the DSS-experiment, no upregulation of chaperone proteins and NfuA were observed, indicating that these are strategies to overcome adverse effects of strong intestinal inflammation. The inhibitor of vertebrate C-type lysozyme, Ivy, was 2- to 3-fold upregulated on mRNA and protein level in E. coli Nissle in comparison to E. coli UNC isolated from IL-10-/- mice. By overexpressing ivy, it was demonstrated in vitro that Ivy contributes to a higher lysozyme resistance observed for E. coli Nissle, supporting the role of Ivy as a potential fitness factor in this E. coli strain. Conclusions: The results of this PhD-study demonstrate that intestinal bacteria sense even minimal changes in the health status of the host. While some bacterial adaptations to the inflammatory conditions are equal in response to strong and mild intestinal inflammation, other reactions are unique to a specific disease state. In addition, probiotic and colitogenic E. coli differ in their response to the intestinal inflammation and thereby may influence the host in different ways.}, language = {en} } @phdthesis{Mabrok2013, author = {Mabrok, Hoda Hussein Bakr}, title = {Protective role of lignan-converting bacteria on chemically-induced breast cancer in gnotobiotic rats}, url = {http://nbn-resolving.de/urn:nbn:de:kobv:517-opus-64933}, school = {Universit{\"a}t Potsdam}, year = {2013}, abstract = {Enterolignans (enterodiol and enterolactone) exhibit structural similarity to estradiol and have therefore been hypothesized to modulate hormone related cancers such as breast cancer. The bioactivation of the plant lignan secoisolariciresinol diglucoside (SDG) requires the transformation by intestinal bacteria including the deglycosylation of SDG to secoisolariciresinol (SECO) followed by demethylation and dehydroxylation of SECO to enterodiol (ED). Finally, ED is dehydrogenated to enterolactone (EL). It is unclear whether the bacterial activation of SDG to ED and EL is crucial for the cancer preventing effects of dietary lignans. The possible protective effect of bacterial lignan transformation on a 7,12 dimethylbenz(a)anthracene (DMBA)-induced breast cancer in gnotobiotic rats was investigated. Germ-free rats were associated with a defined lignan-converting consortium (Clostridium saccharogumia, Blautia producta, Eggerthella lenta, and Lactonifactor longoviformis). The rats colonized with lignan-converting bacteria consortium (LCC) were fed a lignan-rich flaxseed diet and breast cancer was chemical induced. Identically treated germ-free rats served as control. All bacteria of the consortium successfully colonized the intestine of the LCC rats. The plant lignan SDG was converted into the enterolignans ED and EL in the LCC rats but not in the germ-free rats. This transformation did not influence cancer incidence but significantly decreased tumor numbers per tumor-bearing rat, and tumor size. Cell proliferation was significantly inhibited and apoptosis was significantly induced in LCC rats. No differences between LCC and control rats were observed in the expression of the genes encoding the estrogen receptors (ERα and ERβ) and G-coupled protein receptor 30 (GPR30). Similar findings were observed for both insulin-like growth factor 1 (IGF-1) and epidermal growth factor receptor (EGFR) genes involved in tumor growth. Proteome analysis revealed that 24 proteins were differentially expressed in tumor tissue from LCC and germ-free. RanBP-type and C3HC4-type zinc finger-containing protein 1 (RBCK1) and poly(rC)-binding protein 1 (PBCP1) were down-regulated by 3.2- and 2.0-fold, respectively. These proteins are associated with cell proliferation. The activity of selected enzymes involved in the degradation of oxidants in plasma and liver was significantly increased in the LCC rats. However, plasma and liver concentrations of reduced glutathione (non-enzymatic antioxidant) and malondialdehyde (oxidative stress marker) did not differ between the groups. In conclusion, the bacterial conversion of plant lignan to enterolignans beneficially influences their anti-cancer effect. However, the mechanisms involved in these effects remain elusive.}, language = {en} } @phdthesis{Rothe2013, author = {Rothe, Monique}, title = {Response of intestinal Escherichia coli to dietary factors in the mouse intestine}, url = {http://nbn-resolving.de/urn:nbn:de:kobv:517-opus-66387}, school = {Universit{\"a}t Potsdam}, year = {2013}, abstract = {Diet is a major force influencing the intestinal microbiota. This is obvious from drastic changes in microbiota composition after a dietary alteration. Due to the complexity of the commensal microbiota and the high inter-individual variability, little is known about the bacterial response at the cellular level. The objective of this work was to identify mechanisms that enable gut bacteria to adapt to dietary factors. For this purpose, germ-free mice monoassociated with the commensal Escherichia coli K-12 strain MG1655 were fed three different diets over three weeks: a diet rich in starch, a diet rich in non-digestible lactose and a diet rich in casein. Two dimensional gel electrophoresis and electrospray tandem mass spectrometry were applied to identify differentially expressed proteins of E. coli recovered from small intestine and caecum of mice fed the lactose or casein diets in comparison with those of mice fed the starch diet. Selected differentially expressed bacterial proteins were characterised in vitro for their possible roles in bacterial adaptation to the various diets. Proteins belonging to the oxidative stress regulon oxyR such as alkyl hydroperoxide reductase subunit F (AhpF), DNA protection during starvation protein (Dps) and ferric uptake regulatory protein (Fur), which are required for E. coli's oxidative stress response, were upregulated in E. coli of mice fed the lactose-rich diet. Reporter gene analysis revealed that not only oxidative stress but also carbohydrate-induced osmotic stress led to the OxyR-dependent expression of ahpCF and dps. Moreover, the growth of E. coli mutants lacking the ahpCF or oxyR genes was impaired in the presence of non-digestible sucrose. This indicates that some OxyR-dependent proteins are crucial for the adaptation of E. coli to osmotic stress conditions. In addition, the function of two so far poorly characterised E. coli proteins was analysed: 2 deoxy-D gluconate 3 dehydrogenase (KduD) was upregulated in intestinal E. coli of mice fed the lactose-rich diet and this enzyme and 5 keto 4 deoxyuronate isomerase (KduI) were downregulated on the casein-rich diet. Reporter gene analysis identified galacturonate and glucuronate as inducers of the kduD and kduI gene expression. Moreover, KduI was shown to facilitate the breakdown of these hexuronates, which are normally degraded by uronate isomerase (UxaC), altronate oxidoreductase (UxaB), altronate dehydratase (UxaA), mannonate oxidoreductase (UxuB) and mannonate dehydratase (UxuA), whose expression was repressed by osmotic stress. The growth of kduID-deficient E. coli on galacturonate or glucuronate was impaired in the presence of osmotic stress, suggesting KduI and KduD to compensate for the function of the regular hexuronate degrading enzymes under such conditions. This indicates a novel function of KduI and KduD in E. coli's hexuronate metabolism. Promotion of the intracellular formation of hexuronates by lactose connects these in vitro observations with the induction of KduD on the lactose-rich diet. Taken together, this study demonstrates the crucial influence of osmotic stress on the gene expression of E. coli enzymes involved in stress response and metabolic processes. Therefore, the adaptation to diet-induced osmotic stress is a possible key factor for bacterial colonisation of the intestinal environment.}, language = {en} }