Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 598: 40-46, 2022 04 02.
Article in English | MEDLINE | ID: mdl-35151202

ABSTRACT

Hepatocytes injury caused by cytolethal distending toxin (CDT) are major events during helicobacter hepaticus (H.hepaticus) infection. Recent study showed that pre-survival autophagy was promoted against CdtB subunit induced DNA damage. In the present study, we demonstrated that inflammatory cytokines IL-6, IL-1ß, TNF-α, IFN-α, IFN-γ expression and STAT phosphorylation were promoted by CdtB. Besides, CdtB decreased cell viability while promote apoptosis in mouse liver (AML12) cells. Especially, apoptotic protein caspase-9, caspase-3 and PARP were activated while the ratio of Bcl-2/Bax was decreased after CdtB treatment. Moreover, apoptosis induced by CdtB was inhibited due to Erk/p38 MAPK signaling pathway suppression performed with SB203580 or U0126. Meanwhile, we found that CdtB increased autophagic marker levels accompanied by Akt/mTOR/P70S6K signaling pathway in a dose dependent manner. To assess the correlation between autophagy and apoptosis induced by H.hepaticus, chloroquine (CQ, 50 µM) was employed to inhibit autophagy. The result showed that inhibition of autophagy with CQ treatment promoted apoptosis induced by CdtB. Altogether, all these results suggest that CdtB triggers apoptosis via MAPK/Erk/p38 signaling pathway in caspase dependent manner, which was prevented by autophagy in AML12 cells. Collectively, our findings provide new insights into the virulence potential of CdtB on the molecular pathogenesis throughout H.hepaticus infection.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Bacterial Toxins/toxicity , Hepatocytes/drug effects , Hepatocytes/pathology , Animals , Apoptosis/physiology , Autophagy/physiology , Caspases/genetics , Caspases/metabolism , Cell Line , Cytokines/genetics , Gene Expression Regulation/drug effects , Helicobacter hepaticus/pathogenicity , Hepatocytes/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice
2.
Gut Microbes ; 13(1): 1-20, 2021.
Article in English | MEDLINE | ID: mdl-33550886

ABSTRACT

Gut microbiota and the immune system are in constant exchange shaping both host immunity and microbial communities. Here, improper immune regulation can cause inflammatory bowel disease (IBD) and colitis. Antibody therapies blocking signaling through the CD40-CD40L axis showed promising results as these molecules are deregulated in certain IBD patients. To better understand the mechanism, we used transgenic DC-LMP1/CD40 animals with a constitutive CD40-signal in CD11c+ cells, causing a lack of intestinal CD103+ dendritic cells (DCs) and failure to induce regulatory T (iTreg) cells. These mice rapidly develop spontaneous fatal colitis, accompanied by dysbiosis and increased inflammatory IL-17+IFN-γ+ Th17/Th1 and IFN-γ + Th1 cells. In the present study, we analyzed the impact of the microbiota on disease development and detected elevated IgA- and IgG-levels in sera from DC-LMP1/CD40 animals. Their serum antibodies specifically bound intestinal bacteria, and by proteome analysis, we identified a 60 kDa chaperonin GroEL (Hsp60) from Helicobacter hepaticus (Hh) as the main specific antigen targeted in the absence of iTregs. When re-derived to a different Hh-free specific-pathogen-free (SPF) microbiota, mice showed few signs of disease, normal microbiota, and no fatality. Upon recolonization of mice with Hh, the disease developed rapidly. Thus, the present work identifies GroEL/Hsp60 as a major Hh-antigen and its role in disease onset, progression, and outcome in this colitis model. Our results highlight the importance of CD103+ DC- and iTreg-mediated immune tolerance to specific pathobionts to maintain healthy intestinal balance.


Subject(s)
Chaperonin 60/immunology , Colitis/microbiology , Helicobacter hepaticus/pathogenicity , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Colitis/immunology , Dendritic Cells/immunology , Helicobacter hepaticus/immunology , Integrin alpha Chains/immunology , Intestines/immunology , Intestines/microbiology , Mice , Mice, Transgenic , Specific Pathogen-Free Organisms , T-Lymphocytes, Regulatory/immunology
3.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255175

ABSTRACT

The prevalence of gastric Helicobacter pylori (Hp) infection is ~50% of the world population. However, how Hp infection influences inflammatory bowel disease in humans is not fully defined. In this study, we examined whether co-infection with Hp influenced Helicobacter hepaticus (Hh)-induced intestinal pathology in Rag2-/- mice. Rag2-/- mice of both sexes were infected with Hh, of which a subgroup was followed by infection with Hp two weeks later. Co-infected males, but not females, had significantly higher total colitis index scores in the colon at both 10 and 21 weeks post-Hh infection (WPI) and developed more severe dysplasia at 21 WPI compared with mono-Hh males. There were no significant differences in colonization levels of gastric Hp and colonic Hh between sexes or time-points. In addition, mRNA levels of colonic Il-1ß, Ifnγ, Tnfα, Il-17A, Il-17F, Il-18, and Il-23, which play important roles in the development and function of proinflammatory innate lymphoid cell groups 1 and 3, were significantly up-regulated in the dually infected males compared with mono-Hh males at 21 WPI. These data suggest that concomitant Hp infection enhances the inflammatory responses in the colon of-Hh-infected Rag2-/- males, which results in more severe colitis and dysplasia.


Subject(s)
Colitis/genetics , DNA-Binding Proteins/genetics , Helicobacter Infections/genetics , Sex Characteristics , Animals , Coinfection/genetics , Coinfection/microbiology , Colitis/microbiology , Colitis/pathology , Female , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter hepaticus/pathogenicity , Helicobacter pylori/pathogenicity , Humans , Male , Mice , Mice, Knockout
4.
Biochem Biophys Res Commun ; 525(3): 654-661, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32122655

ABSTRACT

It has been documented that Helicobacter hepaticus (H. hepaticus) infection is linked to hepatic inflammation and fibrosis. Interleukin 33 (IL-33) is a cytokine involved in inflammatory and fibrotic diseases, but its relevance to H. hepaticus infection-induced liver inflammation and fibrosis is unknown. In this study, we found that the expression of IL-33 in mice liver was significantly induced by H. hepaticus infection at 24 weeks post infection (WPI). Immunohistochemistry analysis revealed that IL-33 was transferred from the nucleus to the cytoplasm due to infection. The quantitation of inflammatory cytokine and histopathology evaluation showed that IL-33 knockdown attenuated the H. hepaticus-induced hepatic inflammation and fibrosis. More importantly, H. hepaticus promoted the expression of the IL-33 receptor ST2 on cell surfaces, and the expression of ST2 then activated the expression nuclear factor-κB (p65), α-SMA, and Erk1/2. These observations provide novel insights into the pathogenic mechanism of hepatic inflammation and fibrosis during H. hepaticus infection.


Subject(s)
Helicobacter Infections/microbiology , Helicobacter hepaticus/pathogenicity , Inflammation/microbiology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Liver Cirrhosis/microbiology , Liver/pathology , Signal Transduction , Animals , Helicobacter Infections/pathology , Hepatitis, Chronic/complications , Hepatitis, Chronic/microbiology , Hepatitis, Chronic/pathology , Inflammation/complications , Inflammation/pathology , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Male , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Int J Cancer ; 145(4): 1042-1054, 2019 08 15.
Article in English | MEDLINE | ID: mdl-30977112

ABSTRACT

Inflammatory bowel disease and colonic tumors induced by Helicobacter hepaticus (Hh) infection in susceptible mouse strains are utilized to dissect the mechanisms underlying similar human diseases. In our study, infection with genotoxic cytolethal distending toxin-producing Hh in 129/SvEv Rag2-/- Il10-/- gpt delta (RagIl10gpt) mice of both sexes for 21 weeks induced significantly more severe cecal and colonic pathology compared to uninfected controls. The mutation frequencies in the infected RagIl10gpt males were 2.1-fold higher for the cecum and 1.7-fold higher for the colon than male RagIl10gpt controls. In addition, there was a 12.5-fold increase of G:C-to-T:A transversions in the colon of Hh-infected males compared to controls. In contrast, there was no statistical significance in mutation frequencies between infected female Rag2Il10gpt mice and controls. Moreover, Hh infection in RagIl10gpt males significantly up-regulated transcription of Tnfα and iNos, and decreased mRNA levels of cecal Atm compared to the infected females; there was no significant difference in mRNA levels of Il-22, Il-17A, Ifnγ and Atr between the infected males and females. Significantly higher levels of cecal and colonic iNos expression and γH2AX-positive epithelial cells (a biomarker for double-strand DNA breaks [DSB]) in Hh-infected Rag2Il10gpt males vs. Hh-infected females were noted. Finally, Hh infection and associated inflammation increased levels of intestinal mucosa-associated genotoxic colibactin-producing pks+ Escherichia coli. Elevated Tnfα and iNos responses and bacterial genotoxins, in concert with suppression of the DSB repair responses, may have promoted mutagenesis in the lower bowel mucosa of Hh-infected male RagIl10gpt mice.


Subject(s)
Colon/microbiology , DNA-Binding Proteins/genetics , Helicobacter Infections/genetics , Helicobacter hepaticus/pathogenicity , Interleukin-10/genetics , Intestinal Mucosa/microbiology , Mutagenesis/genetics , Animals , Epithelial Cells/microbiology , Female , Helicobacter Infections/microbiology , Inflammation/genetics , Inflammation/microbiology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Interleukin-17/genetics , Male , Mice , Mutation/genetics , RNA, Messenger/genetics , Sex Factors , Tumor Necrosis Factor-alpha/genetics
6.
Mol Hum Reprod ; 25(2): 88-99, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30445548

ABSTRACT

STUDY QUESTION: Does the genotype of the surrogate mother modulate the body composition and immunity of her offspring? SUMMARY ANSWER: C57BL/6J (B6) progenies carried by immunodeficient NOD SCID (NS) mothers had increased adaptive but decreased innate, immune responsiveness in comparison with the same genotype offspring carried by immunocompetent mothers, B6 and BALB/c (C); the B6 progenies carried by the same genotype mothers also showed higher body fat than the others. WHAT IS KNOWN ALREADY: Differences in the major histocompatibility complex (MHC) genes between mother and foetus is considered as an important factor in prenatal embryo development, whereas the impact of such dissimilarity on the phenotype of the mature progeny is unclear. STUDY DESIGN, SIZE, DURATION: Transplantation of two-cell mouse embryos into recipient females of the different MHC (H2) genotypes was used as an approach to simulate three variants of the immunogenic mother-foetus interaction: (i) bidirectional immunogenic dialogue between B6 (H2b haplotype) embryos and C (H2d haplotype) surrogate mother; (ii) one-way immunogenic interaction between B6 embryos and immunodeficient NS (H2g7 haplotype) surrogate mother and (iii) reduced immunogenetic dialogue between embryos and surrogate mother of the same H2b haplotype resulting in only a maternal response to HY antigens of male foetuses. Delivered by Caesarean section, pups were fostered by lactating B6 females and weighed after weaning (n = 171). Body mass and composition and innate and adaptive immunity were assessed in selected progeny groups at 9-11 weeks of age. PARTICIPANTS/MATERIALS, SETTING, METHODS: The study was performed on the specific pathogen-free mouse, inbred strains C57BL/6J, NOD SCID and BALB/c. Plasma progesterone in pregnant females was measured by enzyme-linked immunosorbent assay (ELISA). Body composition was determined by magnetic resonance spectroscopy using a low-field NMR spectrometer (EchoMRI, USA). To assess peritoneal macrophage responses (innate immunity) to anthrax, lactate dehydrogenase (LDH) and interleukin-1 (IL-1ß) were measured in a culture medium 24 h after the addition of both anthrax-lethal factor and anthrax-protective antigen. To assess adaptive immunity, 9-10 males in experimental groups were infected with Helicobacter hepaticus. Faeces collected 2 and 4 weeks after infection was used for quantitative assessment of the H. hepaticus DNA by real-time polymerase chain reaction. IgA, interferon (IFN-γ), tumour necrosis factor (TNFα), interleukin-17 (IL-17) and interleukin-10 (IL-10) in colon tissue and IgG in serum were determined in samples collected 4 weeks after gavage with H. hepaticus using ELISA. For statistical analyses, ANCOVA, post hoc least significant difference (LSD) test, Student's t-test, Spearman rank correlations and χ2 test were performed. P-value <0.05 was considered as a statistically significant difference. MAIN RESULTS AND THE ROLE OF CHANCE: ANCOVA with litter size and age as covariates revealed significant effects of the surrogate mother genotype on body mass and percent of fat in their adult progeny (F2149 = 15.60, P < 0.001 and F2149 = 5.02, P = 0.007, respectively). Adult B6 mice carried by B6 surrogate mothers were characterized by a higher percentage of body fat in comparison with offspring that were carried by NS and C females. In comparison with the male offspring carried by the B6 and C mothers, male B6 progenies carried by immunodeficient NS mothers had a higher humoral immune response (serum IgG) against oral infection with H. hepaticus, but lower in vitro macrophage IL-1ß reaction to the anthrax. Four weeks after the infection of offspring, concentrations of serum IgG and colon IL-10 correlated positively with maternal progesterone on Day 4 after embryo transfer and negatively with DNA of H. hepaticus. One-way ANOVA confirmed a statistically significant impact of surrogate mother genotype on adaptive (IgG) and innate (IL-1ß) immunity (F2.26 = 26.39, P < 0.001 and F2.27 = 5.89, P = 0.008, respectively). LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: The main limitation of our study is the number of combinations of mother and foetus interactions, in particular, transfer of only one embryo genotype was used. Also, it is a descriptive study, which requires further analysis of the epigenetic mechanisms of the observed phenotypic effects of surrogate mother genotype. WIDER IMPLICATIONS OF THE FINDINGS: Our experimental data demonstrate that the transfer of inbred embryos to surrogate mothers of the different genotypes is a prospective experimental model for the study of epigenetic effects of the immunogenetic interactions between mother and foetus. The experimental approach tested in our study will be in demand for the development of criteria for choosing surrogate mothers. In particular, immunocompetence of the surrogate mother along with genetic distance of her MHC alleles to the transferred embryos have a significant impact on offspring development. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the Russian FPI (6/099/2017), budget projects (0324-2016-0002 and 0324-2018-0016) and implemented using the equipment of the Centre for Genetic Resources of Laboratory Animals at ICG SB RAS, supported by the Ministry of Education and Science of Russia (Unique project identifier RFMEFI62117X0015). The authors report no conflicts of interest.


Subject(s)
Embryo Transfer , Embryo, Mammalian/metabolism , Adaptive Immunity/genetics , Adaptive Immunity/physiology , Animals , Anthrax/immunology , Body Composition/physiology , Body Mass Index , Embryo, Mammalian/immunology , Female , Genotype , Helicobacter hepaticus/immunology , Helicobacter hepaticus/pathogenicity , Immunity, Innate/genetics , Immunity, Innate/physiology , Macrophages/immunology , Macrophages/microbiology , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Pregnancy
7.
Nature ; 554(7692): 373-377, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29414937

ABSTRACT

Both microbial and host genetic factors contribute to the pathogenesis of autoimmune diseases. There is accumulating evidence that microbial species that potentiate chronic inflammation, as in inflammatory bowel disease, often also colonize healthy individuals. These microorganisms, including the Helicobacter species, can induce pathogenic T cells and are collectively referred to as pathobionts. However, how such T cells are constrained in healthy individuals is not yet understood. Here we report that host tolerance to a potentially pathogenic bacterium, Helicobacter hepaticus, is mediated by the induction of RORγt+FOXP3+ regulatory T (iTreg) cells that selectively restrain pro-inflammatory T helper 17 (TH17) cells and whose function is dependent on the transcription factor c-MAF. Whereas colonization of wild-type mice by H. hepaticus promoted differentiation of RORγt-expressing microorganism-specific iTreg cells in the large intestine, in disease-susceptible IL-10-deficient mice, there was instead expansion of colitogenic TH17 cells. Inactivation of c-MAF in the Treg cell compartment impaired differentiation and function, including IL-10 production, of bacteria-specific iTreg cells, and resulted in the accumulation of H. hepaticus-specific inflammatory TH17 cells and spontaneous colitis. By contrast, RORγt inactivation in Treg cells had only a minor effect on the bacteria-specific Treg and TH17 cell balance, and did not result in inflammation. Our results suggest that pathobiont-dependent inflammatory bowel disease is driven by microbiota-reactive T cells that have escaped this c-MAF-dependent mechanism of iTreg-TH17 homeostasis.


Subject(s)
Colitis/immunology , Colitis/microbiology , Helicobacter hepaticus/immunology , Immune Tolerance , Intestines/immunology , Intestines/microbiology , Proto-Oncogene Proteins c-maf/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Bioengineering , Colitis/pathology , Female , Forkhead Transcription Factors/metabolism , Helicobacter hepaticus/genetics , Helicobacter hepaticus/pathogenicity , Homeostasis , Host-Pathogen Interactions , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Interleukin-10/immunology , Male , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proto-Oncogene Proteins c-maf/deficiency , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/cytology , Th17 Cells/immunology
8.
Cell Host Microbe ; 23(2): 191-202.e4, 2018 Feb 14.
Article in English | MEDLINE | ID: mdl-29358084

ABSTRACT

Genome-wide association studies have linked polymorphisms in the autophagy gene ATG16L1 with susceptibility to inflammatory bowel disease (IBD). However, the cell-type-specific effects of autophagy on the regulation of chronic intestinal inflammation have not been investigated. Here, we assessed the effect of myeloid-specific or intestinal epithelial cell (IEC)-specific deletion of Atg16l1 on chronic colitis triggered by the intestinal opportunistic pathogen Helicobacter hepaticus in mice. Although Atg16l1 deficiency in myeloid cells had little effect on disease, mice selectively lacking Atg16l1 in IECs (Atg16l1VC) developed severely exacerbated pathology, accompanied by elevated pro-inflammatory cytokine secretion and increased IEC apoptosis. Using ex vivo IEC organoids, we demonstrate that autophagy intrinsically controls TNF-induced apoptosis and in vivo blockade of TNF attenuated the exacerbated pathology in Atg16l1VC mice. These findings suggest that the IBD susceptibility gene ATG16L1 and the process of autophagy within the epithelium control inflammation-induced apoptosis and barrier integrity to limit chronic intestinal inflammation.


Subject(s)
Apoptosis/immunology , Autophagy/immunology , Carrier Proteins/genetics , Colitis/immunology , Goblet Cells/immunology , Paneth Cells/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/genetics , Autophagy/genetics , Autophagy-Related Proteins , Cell Line , Citrobacter rodentium/pathogenicity , HEK293 Cells , Helicobacter hepaticus/pathogenicity , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology
9.
Infect Immun ; 86(2)2018 02.
Article in English | MEDLINE | ID: mdl-29203542

ABSTRACT

Cells of the monocyte/macrophage lineage play important roles in the pathogenesis of inflammatory bowel diseases, but they are also present in the normal healthy intestine, where they are critical for maintaining homeostasis. It has been unclear whether the proinflammatory roles of intestinal macrophages reflect altered behavior of the existing resident cells, or whether they involve recruitment of a distinct cell type. Here, we have explored these ideas using the model of colitis induced by Helicobacter hepaticus in the context of neutralization or deletion of interleukin-10 (IL-10). Granulocytes and monocytes made up most of the inflammatory myeloid infiltrates found in the colon of H. hepaticus-infected colitic mice, rising to a peak within 2 weeks of H. hepaticus inoculation but taking several months to resolve completely. The inflammatory response was dependent on the combined presence of H. hepaticus and absence of IL-10 and was accompanied by increased production of inflammatory mediators such as IL-1ß, tumor necrosis factor alpha (TNF-α), IL-6, and IL-23p19 by infiltrating myeloid cells, mostly relatively immature cells of the macrophage lineage that express intermediate levels of CX3CR1. In contrast, the population of mature CX3CR1hi macrophages did not expand as markedly during colitis, and these cells made little contribution to inflammatory mediator production. Taking into account their numerical dominance in the myeloid compartment, we conclude that newly recruited monocytes are the main source of proinflammatory mediators in colitis induced in the absence of IL-10 signaling and that altered behavior of mature macrophages is not a major component of this pathology.


Subject(s)
CX3C Chemokine Receptor 1/analysis , Colitis/pathology , Cytokines/metabolism , Helicobacter Infections/pathology , Helicobacter hepaticus/pathogenicity , Macrophages/chemistry , Macrophages/immunology , Animals , Colon/pathology , Disease Models, Animal , Female , Granulocytes/immunology , Mice, Inbred C57BL
10.
Cell Microbiol ; 19(7)2017 07.
Article in English | MEDLINE | ID: mdl-28111881

ABSTRACT

Multiple pathogenic Gram-negative bacteria produce the cytolethal distending toxin (CDT) with activity of DNase I; CDT can induce DNA double-strand breaks (DSBs), G2/M cell cycle arrest, and apoptosis in cultured mammalian cells. However, the link of CDT to in vivo tumorigenesis is not fully understood. In this study, 129/SvEv Rag2-/- mice were gavaged with wild-type Helicobacter hepatics 3B1(Hh) and its isogenic cdtB mutant HhcdtBm7, followed by infection for 10 and 20 weeks (WPI). HhCDT deficiency did not affect cecal colonization levels of HhcdtBm7, but attenuated severity of cecal pathology in HhcdtBm7-infected mice. Of importance, preneoplasic dysplasia was progressed to cancer from 10 to 20 WPI in the Hh-infected mice but not in the HhcdtBm7-infected mice. In addition, the loss of HhCDT significantly dampened transcriptional upregulation of cecal Tnfα and Il-6, but elevated Il-10 mRNA levels when compared to Hh at 10 WPI. Furthermore, the presence of HhCDT increased numbers of lower bowel intestinal γH2AX-positive epithelial cells (a marker of DSBs) at both 10 and 20 WPI and augmented phospho-Stat3 foci+ intestinal crypts (activation of Stat3) at 20 WPI. Our findings suggest that CDT promoted Hh carcinogenesis by enhancing DSBs and activation of the Tnfα/Il-6-Stat3 signaling pathway.


Subject(s)
Bacterial Toxins/metabolism , Carcinogenesis/pathology , DNA Breaks, Double-Stranded , Helicobacter hepaticus/pathogenicity , Interleukin-6/metabolism , Intestines/pathology , STAT3 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Bacterial Toxins/genetics , Cecum/microbiology , Cecum/pathology , Female , G2 Phase Cell Cycle Checkpoints , Helicobacter hepaticus/genetics , Histones/metabolism , Interleukin-10/genetics , Male , Mice , Mice, Transgenic , Neoplasms/microbiology , Neoplasms/pathology , RNA, Messenger/biosynthesis , Signal Transduction/physiology
11.
Oncotarget ; 7(22): 32015-30, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27027442

ABSTRACT

Although IL-10 promotes a regulatory phenotype of CD11c+ dendritic cells and macrophages in vitro, the role of IL-10 signaling in CD11c+ cells to maintain intestinal tolerance in vivo remains elusive. To this aim, we generated mice with a CD11c-specific deletion of the IL-10 receptor alpha (Cd11ccreIl10rafl/fl). In contrast to the colon, the small intestine of Cd11ccreIl10rafl/fl mice exhibited spontaneous crypt hyperplasia, increased numbers of intraepithelial lymphocytes and lamina propria T cells, associated with elevated levels of T cell-derived IFNγ and IL-17A. Whereas naive mucosal T-cell priming was not affected and oral tolerance to ovalbumin was intact, augmented T-cell function in the lamina propria was associated with elevated numbers of locally dividing T cells, expression of T-cell attracting chemokines and reduced T-cell apoptosis. Upon stimulation, intestinal IL-10Rα deficient CD11c+ cells exhibited increased activation associated with enhanced IL-6 and TNFα production. Following colonization with Helicobacter hepaticus Cd11ccreIl10rafl/fl mice developed severe large intestinal inflammation characterized by infiltrating T cells and increased levels of Il17a, Ifng, and Il12p40. Altogether these findings demonstrate a critical role of IL-10 signaling in CD11c+ cells to control small intestinal immune homeostasis by limiting reactivation of local memory T cells and to protect against Helicobacter hepaticus-induced colitis.


Subject(s)
CD11c Antigen/metabolism , Colitis/prevention & control , Helicobacter Infections/prevention & control , Immunity, Mucosal , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Intestine, Large/metabolism , Intestine, Small/metabolism , T-Lymphocytes/metabolism , Animals , CD11c Antigen/deficiency , CD11c Antigen/genetics , CD11c Antigen/immunology , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Disease Models, Animal , Genetic Predisposition to Disease , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter hepaticus/immunology , Helicobacter hepaticus/pathogenicity , Homeostasis , Immunologic Memory , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10 Receptor alpha Subunit/genetics , Interleukin-10 Receptor alpha Subunit/immunology , Interleukin-10 Receptor alpha Subunit/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestine, Large/immunology , Intestine, Large/microbiology , Intestine, Small/immunology , Intestine, Small/microbiology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/microbiology
12.
J Immunol ; 196(1): 34-8, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26590314

ABSTRACT

IL-1 family members are central mediators of host defense. In this article, we show that the novel IL-1 family member IL-36γ was expressed during experimental colitis and human inflammatory bowel disease. Germ-free mice failed to induce IL-36γ in response to dextran sodium sulfate (DSS)-induced damage, suggesting that gut microbiota are involved in its induction. Surprisingly, IL-36R-deficient (Il1rl2(-/-)) mice exhibited defective recovery following DSS-induced damage and impaired closure of colonic mucosal biopsy wounds, which coincided with impaired neutrophil accumulation in the wound bed. Failure of Il1rl2(-/-) mice to recover from DSS-induced damage was associated with a profound reduction in IL-22 expression, particularly by colonic neutrophils. Defective recovery of Il1rl2(-/-) mice could be rescued by an aryl hydrocarbon receptor agonist, which was sufficient to restore IL-22 expression and promote full recovery from DSS-induced damage. These findings implicate the IL-36/IL-36R axis in the resolution of intestinal mucosal wounds.


Subject(s)
Colitis/immunology , Interleukin-1/biosynthesis , Interleukins/biosynthesis , Receptors, Interleukin/immunology , Wound Healing/immunology , Animals , Colitis/chemically induced , Colitis/microbiology , Colon/immunology , Colon/injuries , Dextran Sulfate , Helicobacter hepaticus/pathogenicity , Humans , Inflammation/immunology , Inflammation/pathology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Aryl Hydrocarbon/agonists , Receptors, Interleukin/genetics , Wound Healing/genetics , Interleukin-22
13.
Int Rev Immunol ; 35(1): 67-82, 2016.
Article in English | MEDLINE | ID: mdl-25970037

ABSTRACT

In space, the lifestyle, relative sterility of spaceship and extreme environmental stresses, such as microgravity and cosmic radiation, can compromise the balance between human body and human microbiome. An astronaut's body during spaceflight encounters increased risk for microbial infections and conditions because of immune dysregulation and altered microbiome, i.e. dysbiosis. This risk is further heightened by increase in virulence of pathogens in microgravity. Health status of astronauts might potentially benefit from maintaining a healthy microbiome by specifically managing their diet on space in addition to probiotic therapies. This review focuses on the current knowledge/understanding of how spaceflight affects human immunity and microbiome.


Subject(s)
Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Immunity/radiation effects , Space Flight , Weightlessness/adverse effects , Astronauts , Bacteroides/immunology , Bacteroides/radiation effects , Candida albicans/immunology , Candida albicans/pathogenicity , Clostridiales/immunology , Clostridiales/pathogenicity , Clostridioides difficile/immunology , Clostridioides difficile/pathogenicity , Cosmic Radiation/adverse effects , Cytokines/immunology , Cytokines/metabolism , Cytokines/radiation effects , Dendritic Cells/metabolism , Dendritic Cells/radiation effects , Dietary Supplements , Escherichia coli/immunology , Escherichia coli/pathogenicity , Gastrointestinal Microbiome/radiation effects , Helicobacter hepaticus/immunology , Helicobacter hepaticus/pathogenicity , Humans , Leukocytes/metabolism , Leukocytes/radiation effects , Probiotics/therapeutic use , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Virulence
14.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G338-46, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24875104

ABSTRACT

Adenosine is a purine metabolite that can mediate anti-inflammatory responses in the digestive tract through the A(2A) adenosine receptor (A(2A)AR). We examined the role of this receptor in the control of inflammation in the adoptive transfer model of colitis. Infection of A(2A)AR(-/-) mice with Helicobacter hepaticus increased colonic inflammation scores compared with uninfected A(2A)AR controls. Comparison of T cell subsets in wild-type and A(2A)AR(-/-) mice revealed differences in markers associated with activated helper T (Th) cells and regulatory T (Treg) cells. Previous studies showed that expression of A(2A)AR on CD45RB(HI) and CD45RB(LO) Th cells is essential for the proper regulation of colonic inflammation. Adoptive transfer of CD45RB(HI) with CD45RB(LO) from wild-type mice into RAG1(-/-)/A(2A)AR(-/-) mice induced severe disease within 3 wk, although transfer of the same subsets into RAG1(-/-) mice does not induce colitis. This suggests that the presence of A(2A)AR on recipient cells is also important for controlling colitis. To investigate the role of A(2A)AR in myeloid cells, chimeric recipients were generated by injection of bone marrow from RAG1(-/-) or RAG1(-/-)/A(2A)AR(-/-) mice into irradiated RAG1(-/-) mice. After adoptive transfer, these recipients did not develop colitis, regardless of A(2A)AR expression by the donor. Together, our results suggest that the control of inflammation in vivo is dependent on A(2A)AR signaling through multiple cell types that collaborate in the regulation of colitis by responding to extracellular adenosine.


Subject(s)
Adenosine/metabolism , Colitis/prevention & control , Colon/metabolism , Lymph Nodes/metabolism , T-Lymphocyte Subsets/metabolism , Adoptive Transfer , Animals , Biomarkers/metabolism , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Colon/immunology , Colon/microbiology , Cytokines/metabolism , Disease Models, Animal , Female , Helicobacter hepaticus/pathogenicity , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Inflammation Mediators/metabolism , Leukocyte Common Antigens/metabolism , Lymph Nodes/immunology , Lymph Nodes/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors
15.
PLoS One ; 8(8): e70783, 2013.
Article in English | MEDLINE | ID: mdl-23951007

ABSTRACT

The mouse pathobiont Helicobacter hepaticus can induce typhlocolitis in interleukin-10-deficient mice, and H. hepaticus infection of immunodeficient mice is widely used as a model to study the role of pathogens and commensal bacteria in the pathogenesis of inflammatory bowel disease. C57BL/6J Il10(-/-) mice kept under specific pathogen-free conditions in two different facilities (MHH and MIT), displayed strong differences with respect to their susceptibilities to H. hepaticus-induced intestinal pathology. Mice at MIT developed robust typhlocolitis after infection with H. hepaticus, while mice at MHH developed no significant pathology after infection with the same H. hepaticus strain. We hypothesized that the intestinal microbiota might be responsible for these differences and therefore performed high resolution analysis of the intestinal microbiota composition in uninfected mice from the two facilities by deep sequencing of partial 16S rRNA amplicons. The microbiota composition differed markedly between mice from both facilities. Significant differences were also detected between two groups of MHH mice born in different years. Of the 119 operational taxonomic units (OTUs) that occurred in at least half the cecum or colon samples of at least one mouse group, 24 were only found in MIT mice, and another 13 OTUs could only be found in MHH samples. While most of the MHH-specific OTUs could only be identified to class or family level, the MIT-specific set contained OTUs identified to genus or species level, including the opportunistic pathogen, Bilophila wadsworthia. The susceptibility to H. hepaticus-induced colitis differed considerably between Il10(-/-) mice originating from the two institutions. This was associated with significant differences in microbiota composition, highlighting the importance of characterizing the intestinal microbiome when studying murine models of IBD.


Subject(s)
Colitis/microbiology , Disease Susceptibility/microbiology , Helicobacter Infections/microbiology , Helicobacter hepaticus/pathogenicity , Interleukin-10/immunology , Microbiota/immunology , Animals , Cecum/immunology , Cecum/microbiology , Cecum/pathology , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/microbiology , Colon/pathology , DNA, Complementary/classification , DNA, Complementary/genetics , Disease Susceptibility/immunology , Helicobacter Infections/immunology , Helicobacter Infections/pathology , Helicobacter hepaticus/physiology , Interleukin-10/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Ribosomal, 16S/classification , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
16.
Inflamm Bowel Dis ; 19(8): 1586-97, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23702805

ABSTRACT

BACKGROUND: Patients with inflammatory bowel disease (IBD) are at increase risk for bone loss and fractures. Therefore, in the present study, we examined the effect of experimental IBD on bone health. METHODS: We used a murine model of colitis, Helicobacter hepaticus-infected interleukin-10-deficient animals. Molecular and histological properties of bone and intestine were examined to identify the immunopathological consequences of colitis in male and female mice. RESULTS: At 6 weeks postinfection, we observed significant trabecular bone loss in male mice but surprisingly not in female mice. This was true for both distal femur and vertebral locations. In addition, H. hepaticus infection suppressed osteoblast markers only in male mice. Consistent with effects on bone health, male mice with H. hepaticus infection had more severe colitis as determined by histology and elevated levels of inflammatory cytokines in the colon. Although H. hepaticus levels in the stool appeared similar in male and female mice 1 week after infection, by 6 weeks, H. hepaticus levels were greater in male mice, indicating that H. hepaticus survival and virulence within the gastrointestinal tract could be gender dependent. CONCLUSION: In summary, H. hepaticus-induced colitis severity and associated bone loss is gender regulated, possibly as a result of gender-specific effects on H. hepaticus colonization in the mouse gastrointestinal tract and the consequent immunopathological responses.


Subject(s)
Bone Diseases/etiology , Colitis/complications , Helicobacter Infections/complications , Helicobacter hepaticus/pathogenicity , Inflammation/etiology , Interleukin-10/physiology , Intestines/pathology , Animals , Blotting, Western , Bone Density , Bone Diseases/pathology , Colitis/microbiology , Colitis/pathology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Inflammation/pathology , Intestines/microbiology , Male , Mice , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors
17.
Cell Microbiol ; 15(6): 992-1011, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23278999

ABSTRACT

The enterohepatic Epsilonproteobacterium Helicobacter hepaticus persistently colonizes the intestine of mice and causes chronic inflammatory symptoms in susceptible mouse strains. The bacterial factors causing intestinal inflammation are poorly characterized. A large genomic pathogenicity island, HHGI1, which encodes components of a type VI secretion system (T6SS), was previously shown to contribute to the colitogenic potential of H. hepaticus. We have now characterized the T6SS components Hcp, VgrG1, VgrG2 and VgrG3, encoded on HHGI1, including the potential impact of the T6SS on intestinal inflammation in a mouse T-cell transfer model. The H. hepaticus T6SS components were expressed during the infection and secreted in a T6SS-dependent manner, when the bacteria were cultured either in the presence or in the absence of mouse intestinal epithelial cells. Mutants deficient in VgrG1 displayed a significantly lower colitogenic potential in T-cell-transferred C57BL/6 Rag2(-/-) mice, despite an unaltered ability to colonize mice persistently. Intestinal microbiota analyses demonstrated only minor changes in mice infected with wild-typeH. hepaticus as compared with mice infected with VgrG1-deficient isogenic bacteria. In addition, competitive assays between both wild-type and T6SS-deficient H. hepaticus, and between wild-type H. hepaticus and Campylobacter jejuni or Enterobacteriaceae species did not show an effect of the T6SS on interbacterial competitiveness. Therefore, we suggest that microbiota alterations did not play a major role in the changes of pro-inflammatory potential mediated by the T6SS. Cellular innate pro-inflammatory responses were increased by the secreted T6SS proteins VgrG1 and VgrG2. We therefore concluded that the type VI secretion component VgrG1 can modulate and specifically exacerbate the innate pro-inflammatory effect of the chronic H. hepaticus infection.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems/physiology , Colitis/microbiology , Helicobacter Infections/physiopathology , Helicobacter hepaticus/physiology , Helicobacter hepaticus/pathogenicity , Animals , Bacterial Proteins/physiology , Campylobacter jejuni/physiology , Cells, Cultured , Colitis/metabolism , Colitis/physiopathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Enterobacteriaceae/physiology , Helicobacter Infections/metabolism , Helicobacter hepaticus/genetics , In Vitro Techniques , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics
18.
Helicobacter ; 18(1): 33-40, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23067369

ABSTRACT

BACKGROUND: The heterogeneity of hepatitis C virus (HCV) infection cannot always be explained by HCV genotypes or host genetic factors, raising the issue of possible cofactors. A new form of hepatitis leading to liver cancer was discovered in 1992 in mice, owing to an infection by Helicobacter hepaticus. Moreover, several studies showed an association between the presence of HCV and Helicobacter in the liver of patients with severe liver diseases suggesting a possible synergism between the two pathogens. In an HCV transgenic mouse model with a B6C3F1 background, the combination of H. hepaticus infection and the HCV transgene resulted in a significantly greater incidence and multiplicity of preneoplastic and neoplastic liver foci in males. OBJECTIVES: Because the mouse genetic background is a major determinant in the development of liver disease, our aim was to test the synergism between HCV and H. hepaticus infection using transgenic mice with a more sensitive genetic background to H. hepaticus infection. METHODS: For this purpose, four groups of mice were followed up to 14 months, the presence of H. hepaticus was monitored by PCR and hepatic lesions were looked for. RESULTS: We found that H. hepaticus, but not the HCV transgene, increased the number of hepatic lesions. The presence of carcinoma was more likely to occur on a background of hepatitis, and the overall lesions were more frequent in the presence of steatosis. The effect of the mouse genetic background was greater than the effect of the HCV transgene and was sufficient to promote lesions particularly via its sensitivity to H. hepaticus infection. CONCLUSIONS: Genetic susceptibility may be a more important factor than expected. Indeed, the synergism between HCV and H. hepaticus infection involved in liver disease may be highly host dependent.


Subject(s)
Helicobacter Infections/pathology , Helicobacter hepaticus/pathogenicity , Hepacivirus/pathogenicity , Hepatitis C/pathology , Liver/pathology , Animals , Coinfection/microbiology , Coinfection/pathology , Coinfection/virology , Disease Models, Animal , Follow-Up Studies , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Hepatitis C/complications , Hepatitis C/virology , Male , Mice , Mice, Transgenic
19.
J Exp Med ; 209(9): 1595-609, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22891275

ABSTRACT

Although very high levels of interleukin (IL)-1ß are present in the intestines of patients suffering from inflammatory bowel diseases (IBD), little is known about the contribution of IL-1ß to intestinal pathology. Here, we used two complementary models of chronic intestinal inflammation to address the role of IL-1ß in driving innate and adaptive pathology in the intestine. We show that IL-1ß promotes innate immune pathology in Helicobacter hepaticus-triggered intestinal inflammation by augmenting the recruitment of granulocytes and the accumulation and activation of innate lymphoid cells (ILCs). Using a T cell transfer colitis model, we demonstrate a key role for T cell-specific IL-1 receptor (IL-1R) signals in the accumulation and survival of pathogenic CD4(+) T cells in the colon. Furthermore, we show that IL-1ß promotes Th17 responses from CD4(+) T cells and ILCs in the intestine, and we describe synergistic interactions between IL-1ß and IL-23 signals that sustain innate and adaptive inflammatory responses in the gut. These data identify multiple mechanisms through which IL-1ß promotes intestinal pathology and suggest that targeting IL-1ß may represent a useful therapeutic approach in IBD.


Subject(s)
CD5 Antigens/metabolism , Colitis/immunology , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Th17 Cells/metabolism , Animals , Cell Survival , Colitis/metabolism , Colitis/microbiology , Colon/immunology , Colon/microbiology , Colon/pathology , Granulocytes/immunology , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Helicobacter hepaticus/pathogenicity , Immunity, Innate , Interleukin-23/metabolism , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1 Type I/metabolism , Th17 Cells/immunology
20.
World J Gastroenterol ; 18(13): 1459-69, 2012 Apr 07.
Article in English | MEDLINE | ID: mdl-22509077

ABSTRACT

AIM: To identify and characterize drosophila mothers against decapentaplegic (SMAD)3-dependent changes in immune cell populations following infection with Helicobacter hepaticus (H. hepaticus). METHODS: SMAD3(-/-) (n = 19) and colitis-resistant SMAD3(+/-) (n = 24) mice (8-10 wk of age) were infected with H. hepaticus and changes in immune cell populations [T lymphocytes, natural killer (NK) cells, T regulatory cells] were measured in the spleen and mesenteric lymph nodes (MsLNs) at 0 d, 3 d, 7 d and 28 d post-infection using flow cytometry. Genotype-dependent changes in T lymphocytes and granzyme B(+) cells were also assessed after 28 d in proximal colon tissue using immunohistochemistry. RESULTS: As previously observed, SMAD3(-/-), but not SMAD3(+/-) mice, developed colitis, peaking at 4 wk post-infection. No significant changes in T cell subsets were observed in the spleen or in the MsLNs between genotypes at any time point. However, CD4(+) and CD8(+)/CD62L(lo) cells, an effector T lymphocyte population, as well as NK cells (NKp46/DX5(+)) were significantly higher in the MsLNs of SMAD3(-/-) mice at 7 d and 28 d post-infection. In the colon, a higher number of CD3(+) cells were present in SMAD3(-/-) compared to SMAD3(+/-) mice at baseline, which did not significantly change during infection. However, the number of granzyme B(+) cells, a marker of cytolytic lymphocytes, significantly increased in SMAD3(-/-) mice 28 d post-infection compared to both SMAD3(+/-) mice and to baseline values. This was consistent with more severe colitis development in these animals. CONCLUSION: Data suggest that defects in SMAD3 signaling increase susceptibility to H. hepaticus-induced colitis through aberrant activation and/or dysregulation of effector lymphocytes.


Subject(s)
Colitis/immunology , Colitis/microbiology , Helicobacter hepaticus/immunology , Helicobacter hepaticus/pathogenicity , Lymphocytes/immunology , Smad3 Protein/immunology , Animals , Colon/cytology , Colon/immunology , Colon/pathology , Female , Granzymes/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocytes/cytology , Male , Mice , Mice, Knockout , Signal Transduction/immunology , Smad3 Protein/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...