Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Nat Immunol ; 25(5): 886-901, 2024 May.
Article in English | MEDLINE | ID: mdl-38609547

ABSTRACT

Intestinal immune responses to microbes are controlled by the cytokine IL-10 to avoid immune pathology. Here, we use single-cell RNA sequencing of colon lamina propria leukocytes (LPLs) along with RNA-seq and ATAC-seq of purified CD4+ T cells to show that the transcription factors Blimp-1 (encoded by Prdm1) and c-Maf co-dominantly regulate Il10 while negatively regulating proinflammatory cytokines in effector T cells. Double-deficient Prdm1fl/flMaffl/flCd4Cre mice infected with Helicobacter hepaticus developed severe colitis with an increase in TH1/NK/ILC1 effector genes in LPLs, while Prdm1fl/flCd4Cre and Maffl/flCd4Cre mice exhibited moderate pathology and a less-marked type 1 effector response. LPLs from infected Maffl/flCd4Cre mice had increased type 17 responses with increased Il17a and Il22 expression and an increase in granulocytes and myeloid cell numbers, resulting in increased T cell-myeloid-neutrophil interactions. Genes over-expressed in human inflammatory bowel disease showed differential expression in LPLs from infected mice in the absence of Prdm1 or Maf, revealing potential mechanisms of human disease.


Subject(s)
Colitis , Helicobacter hepaticus , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-maf , Animals , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Mice , Proto-Oncogene Proteins c-maf/genetics , Colitis/immunology , Colitis/genetics , Humans , Helicobacter hepaticus/immunology , Helicobacter Infections/immunology , Mice, Inbred C57BL , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/genetics , Gene Expression Regulation , Disease Models, Animal
2.
Nature ; 628(8009): 854-862, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570678

ABSTRACT

The intestinal immune system is highly adapted to maintaining tolerance to the commensal microbiota and self-antigens while defending against invading pathogens1,2. Recognizing how the diverse network of local cells establish homeostasis and maintains it in the complex immune environment of the gut is critical to understanding how tolerance can be re-established following dysfunction, such as in inflammatory disorders. Although cell and molecular interactions that control T regulatory (Treg) cell development and function have been identified3,4, less is known about the cellular neighbourhoods and spatial compartmentalization that shapes microorganism-reactive Treg cell function. Here we used in vivo live imaging, photo-activation-guided single-cell RNA sequencing5-7 and spatial transcriptomics to follow the natural history of T cells that are reactive towards Helicobacter hepaticus through space and time in the settings of tolerance and inflammation. Although antigen stimulation can occur anywhere in the tissue, the lamina propria-but not embedded lymphoid aggregates-is the key microniche that supports effector Treg (eTreg) cell function. eTreg cells are stable once their niche is established; however, unleashing inflammation breaks down compartmentalization, leading to dominance of CD103+SIRPα+ dendritic cells in the lamina propria. We identify and validate the putative tolerogenic interaction between CD206+ macrophages and eTreg cells in the lamina propria and identify receptor-ligand pairs that are likely to govern the interaction. Our results reveal a spatial mechanism of tolerance in the lamina propria and demonstrate how knowledge of local interactions may contribute to the next generation of tolerance-inducing therapies.


Subject(s)
Intestinal Mucosa , Mucous Membrane , T-Lymphocytes, Regulatory , Animals , Female , Male , Mice , Antigens, CD/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Profiling , Helicobacter hepaticus/immunology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Immune Tolerance/immunology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Integrin alpha Chains/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mucous Membrane/cytology , Mucous Membrane/immunology , Receptors, Immunologic/metabolism , Receptors, Immunologic/immunology , Single-Cell Gene Expression Analysis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/cytology , Transcriptome
3.
Immunology ; 164(3): 476-493, 2021 11.
Article in English | MEDLINE | ID: mdl-34322877

ABSTRACT

In recent years, an increasing number of studies have reported that intestinal microbiota have an important effect on tumour immunity by affecting the tumour microenvironment (TME). The intestinal microbiota are closely associated with various immune cells, such as T lymphocytes, natural killer cells (NK cells) and macrophages. Some bacteria, such as Akkermansia muciniphila (A. muciniphila) and Lactobacillus reuteri (L. reuteri), have been shown to improve the effect of tumour immunity. Furthermore, microbial imbalance, such as the increased abundance of Fusobacterium nucleatum (F. nucleatum) and Helicobacter hepaticus (H. hepaticus), generally causes tumour formation and progression. In addition, some microbiota also play important roles in tumour immunotherapy, especially PD-L1-related therapies. Therefore, what is the relationship between these processes and how do they affect each other? In this review, we summarize the interactions and corresponding mechanisms among the intestinal microbiota, immune system and TME to facilitate the research and development of new targeted drugs and provide new approaches to tumour therapy.


Subject(s)
Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Disease Models, Animal , Disease Progression , Dysbiosis/microbiology , Dysbiosis/pathology , Fusobacterium nucleatum/immunology , Gastrointestinal Microbiome/drug effects , Helicobacter hepaticus/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Killer Cells, Natural/immunology , Macrophages/immunology , Neoplasms/drug therapy , Neoplasms/microbiology , Neoplasms/pathology , T-Lymphocytes/immunology , Tumor Microenvironment/drug effects
4.
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
5.
Sci Immunol ; 5(47)2020 05 22.
Article in English | MEDLINE | ID: mdl-32444476

ABSTRACT

Mononuclear phagocytes (MNPs) are vital for maintaining intestinal homeostasis but, in response to acute microbial stimulation, can also trigger immunopathology, accelerating recruitment of Ly6Chi monocytes to the gut. The regulators that control monocyte tissue adaptation in the gut remain poorly understood. Interferon regulatory factor 5 (IRF5) is a transcription factor previously shown to play a key role in maintaining the inflammatory phenotype of macrophages. Here, we investigate the impact of IRF5 on the MNP system and physiology of the gut at homeostasis and during inflammation. We demonstrate that IRF5 deficiency has a limited impact on colon physiology at steady state but ameliorates immunopathology during Helicobacter hepaticus-induced colitis. Inhibition of IRF5 activity in MNPs phenocopies global IRF5 deficiency. Using a combination of bone marrow chimera and single-cell RNA-sequencing approaches, we examined the intrinsic role of IRF5 in controlling colonic MNP development. We demonstrate that IRF5 promotes differentiation of Ly6Chi monocytes into CD11c+ macrophages and controls the production of antimicrobial and inflammatory mediators by these cells. Thus, we identify IRF5 as a key transcriptional regulator of the colonic MNP system during intestinal inflammation.


Subject(s)
CD11 Antigens/immunology , Inflammation/immunology , Interferon Regulatory Factors/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Helicobacter hepaticus/immunology , Inflammation/pathology , Interferon Regulatory Factors/deficiency , Macrophages/pathology , Mice , Mice, Knockout , Monocytes/pathology , Phenotype
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.
Nat Commun ; 9(1): 3797, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30228258

ABSTRACT

Inflammatory bowel disease (IBD) are heterogenous disorders of the gastrointestinal tract caused by a spectrum of genetic and environmental factors. In mice, overlapping regions of chromosome 3 have been associated with susceptibility to IBD-like pathology, including a locus called Hiccs. However, the specific gene that controls disease susceptibility remains unknown. Here we identify a Hiccs locus gene, Alpk1 (encoding alpha kinase 1), as a potent regulator of intestinal inflammation. In response to infection with the commensal pathobiont Helicobacter hepaticus (Hh), Alpk1-deficient mice display exacerbated interleukin (IL)-12/IL-23 dependent colitis characterized by an enhanced Th1/interferon(IFN)-γ response. Alpk1 controls intestinal immunity via the hematopoietic system and is highly expressed by mononuclear phagocytes. In response to Hh, Alpk1-/- macrophages produce abnormally high amounts of IL-12, but not IL-23. This study demonstrates that Alpk1 promotes intestinal homoeostasis by regulating the balance of type 1/type 17 immunity following microbial challenge.


Subject(s)
Colitis/immunology , Helicobacter Infections/immunology , Inflammatory Bowel Diseases/immunology , Interleukin-12/immunology , Protein Kinases/metabolism , Th1 Cells/immunology , Animals , Bone Marrow Cells , Bone Marrow Transplantation , Colitis/microbiology , Colitis/pathology , Colon , Disease Models, Animal , Female , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter hepaticus/immunology , Humans , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Interleukin-12/metabolism , Interleukin-23/immunology , Interleukin-23/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , Protein Kinases/genetics , Protein Kinases/immunology , Radiation Chimera , Th1 Cells/metabolism
8.
Front Immunol ; 9: 499, 2018.
Article in English | MEDLINE | ID: mdl-29636751

ABSTRACT

Chronic inflammation contributes to tumor initiation in colitis-associated colorectal cancer (CRC). Indeed, inflammatory bowel disease (IBD) patients show an increased risk of developing CRC. Cancer immune evasion is a major issue in CRC and preclinical and clinical evidence has defined a critical role for myeloid-derived suppressor cells (MDSCs) that contribute to tumor growth and progression by suppressing T-cells and modulating innate immune responses. MDSCs comprise a heterogeneous population of immature myeloid cells that can be distinct in two subtypes: CD11b+Ly6G+Ly6Clow with granulocytic phenotype (G-MDSCs) and CD11b+Ly6G-Ly6Chigh with monocytic phenotype (M-MDSCs). Hydrogen sulfide (H2S) is an endogenous gaseous signaling molecule that regulates various physiological and pathophysiological functions. In particular, several studies support its anti-inflammatory activity in experimental colitis and ulcer. However, the role of the H2S pathway in innate immune-mediated IBD has not yet been elucidated. To better define a possible link between MDSCs and H2S pathway in colitis-associated CRC development, we used an innate immune-mediated IBD model induced by infection with the bacterium Helicobacter hepaticus (Hh), closely resembling human IBD. Here, we demonstrated an involvement of MDSCs in colitis development. A significant time-dependent increase of both G-MDSCs and M-MDSCs was observed in the colon and in the spleen of Hh-infected mice. Following, we observed that chronic oral administration of the H2S donor DATS reduced colon inflammation by limiting the recruitment of G-MDSCs in the colon of Hh-infected mice. Thus, we identify the metabolic pathway l-cysteine/H2S as a possible new player in the immunosuppressive mechanism responsible for the MDSCs-promoted colitis-associated cancer development.


Subject(s)
Colitis/immunology , Helicobacter Infections/immunology , Helicobacter hepaticus/immunology , Hydrogen Sulfide/pharmacology , Immunity, Cellular/drug effects , Myeloid-Derived Suppressor Cells/immunology , Animals , Colitis/genetics , Colitis/microbiology , Colitis/pathology , Colon/immunology , Colon/microbiology , Colon/pathology , Disease Models, Animal , Helicobacter Infections/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Mice , Mice, Knockout , Myeloid-Derived Suppressor Cells/pathology
9.
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
10.
Cell Host Microbe ; 22(6): 733-745.e5, 2017 Dec 13.
Article in English | MEDLINE | ID: mdl-29241040

ABSTRACT

Interactions between the host and its microbiota are of mutual benefit and promote health. Complex molecular pathways underlie this dialog, but the identity of microbe-derived molecules that mediate the mutualistic state remains elusive. Helicobacter hepaticus is a member of the mouse intestinal microbiota that is tolerated by the host. In the absence of an intact IL-10 signaling, H. hepaticus induces an IL-23-driven inflammatory response in the intestine. Here we investigate the interactions between H. hepaticus and host immune cells that may promote mutualism, and the microbe-derived molecule(s) involved. Our results show that H. hepaticus triggers early IL-10 induction in intestinal macrophages and produces a large soluble polysaccharide that activates a specific MSK/CREB-dependent anti-inflammatory and repair gene signature via the receptor TLR2. These data identify a host-bacterial interaction that promotes mutualistic mechanisms at the intestinal interface. Further understanding of this pathway may provide novel prevention and treatment strategies for inflammatory bowel disease.


Subject(s)
Helicobacter hepaticus/immunology , Helicobacter hepaticus/metabolism , Immunosuppressive Agents/metabolism , Macrophages/drug effects , Macrophages/immunology , Polysaccharides, Bacterial/metabolism , Symbiosis , Animals , Interleukin-10/metabolism , Interleukin-23/metabolism , Mice , Toll-Like Receptor 2/metabolism
12.
Mucosal Immunol ; 10(6): 1504-1517, 2017 11.
Article in English | MEDLINE | ID: mdl-28198364

ABSTRACT

The risk of colon cancer is increased in patients with Crohn's disease and ulcerative colitis. Inflammation-induced DNA damage could be an important link between inflammation and cancer, although the pathways that link inflammation and DNA damage are incompletely defined. RAG2-deficient mice infected with Helicobacter hepaticus (Hh) develop colitis that progresses to lower bowel cancer. This process depends on nitric oxide (NO), a molecule with known mutagenic potential. We have previously hypothesized that production of NO by macrophages could be essential for Hh-driven carcinogenesis, however, whether Hh infection induces DNA damage in this model and whether this depends on NO has not been determined. Here we demonstrate that Hh infection of RAG2-deficient mice rapidly induces expression of iNOS and the development of DNA double-stranded breaks (DSBs) specifically in proliferating crypt epithelial cells. Generation of DSBs depended on iNOS activity, and further, induction of iNOS, the generation of DSBs, and the subsequent development of dysplasia were inhibited by depletion of the Hh-induced cytokine IL-22. These results demonstrate a strong association between Hh-induced DNA damage and the development of dysplasia, and further suggest that IL-22-dependent induction of iNOS within crypt epithelial cells rather than macrophages is a driving force in this process.


Subject(s)
Colitis, Ulcerative/immunology , Colon/pathology , Colonic Neoplasms/immunology , Helicobacter Infections/immunology , Helicobacter hepaticus/immunology , Inflammation/immunology , Interleukins/metabolism , Macrophages, Peritoneal/immunology , Animals , Antibodies, Blocking/administration & dosage , Colitis, Ulcerative/complications , Colon/physiopathology , Colonic Neoplasms/complications , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , Disease Models, Animal , Helicobacter Infections/complications , Humans , Interleukins/immunology , Macrophages, Peritoneal/microbiology , Mice , Mice, 129 Strain , Mice, Knockout , Neoplasms , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Interleukin-22
13.
J Infect ; 73(6): 523-535, 2016 12.
Article in English | MEDLINE | ID: mdl-27746159

ABSTRACT

OBJECTIVES: Although n-3 long-chain polyunsaturated fatty acids (n-3 LC-PUFAs) are used widely in the treatment of chronic inflammatory diseases, their effect in infectious disease requires a particular attention. METHODS: The present article discusses their anti-inflammatory and immune properties involved in the host defence and presents a systematic review of the effects of their oral administration on the prevention and outcome of experimental and clinical infections. RESULTS: At a dose corresponding to an human dose of 500 mg/day, n-3 LC-PUFAs intake is beneficial against experimental infections caused by extracellular pathogens including Streptococcus pneumoniae, Pseudomonas aeruginosa, Escherichia coli, and Staphylococcus aureus by reducing inflammation, and reduces the incidence of pneumococcal infections in the elderly, but at 2-4-fold higher doses as occurs in some human intervention and/or during long-term it becomes detrimental in intestinal infections with Citrobacter rodentium or Helicobacter hepaticus by exacerbating anti-inflammatory response. They are also harmful against infections caused by intracellular pathogens as Mycobacterium tuberculosis, Salmonella, Influenza virus and Herpes simplex virus by affecting the immune cell response. CONCLUSION: The effects of n-3-LC-PUFAs on infections depend on the pathogen and the n-3 LC-PUFA dose and timing. Caution should be recommended for high-dose and long-term supplementation in humans.


Subject(s)
Bacterial Infections/prevention & control , Fatty Acids, Omega-3/administration & dosage , Virus Diseases/prevention & control , Administration, Oral , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Chronic Disease/drug therapy , Chronic Disease/prevention & control , Citrobacter rodentium/drug effects , Citrobacter rodentium/immunology , Dietary Supplements , Fatty Acids, Omega-3/adverse effects , Fatty Acids, Omega-3/therapeutic use , Helicobacter hepaticus/drug effects , Helicobacter hepaticus/immunology , Herpes Simplex/drug therapy , Herpes Simplex/prevention & control , Herpes Simplex/virology , Humans , Inflammation/drug therapy , Inflammation/prevention & control , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/immunology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/prevention & control , Staphylococcus aureus/drug effects , Staphylococcus aureus/immunology , Virus Diseases/drug therapy , Virus Diseases/virology
14.
J Immunol ; 196(12): 4893-904, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27183623

ABSTRACT

The discovery of Th17 cell plasticity, in which CD4(+) IL-17-producing Th17 cells give rise to IL-17/IFN-γ double-producing cells and Th1-like IFNγ(+) ex-Th17 lymphocytes, has raised questions regarding which of these cell types contribute to immunopathology during inflammatory diseases. In this study, we show using Helicobacter hepaticus-induced intestinal inflammation that IL-17A(Cre)- or Rag1(Cre)-mediated deletion of Tbx21 has no effect on the generation of IL-17/IFN-γ double-producing cells, but leads to a marked absence of Th1-like IFNγ(+) ex-Th17 cells. Despite the lack of Th1-like ex-Th17 cells, the degree of H. hepaticus-triggered intestinal inflammation in mice in which Tbx21 was excised in IL-17-producing or Rag1-expressing cells is indistinguishable from that observed in control mice. In stark contrast, using experimental autoimmune encephalomyelitis, we show that IL-17A(Cre)-mediated deletion of Tbx21 prevents the conversion of Th17 cells to IL-17A/IFN-γ double-producing cells as well as Th1-like IFN-γ(+) ex-Th17 cells. However, IL-17A(Cre)-mediated deletion of Tbx21 has only limited effects on disease course in this model and is not compensated by Ag-specific Th1 cells. IL-17A(Cre)-mediated deletion of Rorc reveals that RORγt is essential for the maintenance of the Th17 cell lineage, but not immunopathology during experimental autoimmune encephalomyelitis. These results show that neither the single Th17 subset, nor its progeny, is solely responsible for immunopathology or autoimmunity.


Subject(s)
Enteritis/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Box Domain Proteins/metabolism , Th17 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Helicobacter hepaticus/immunology , Interferon-gamma/biosynthesis , Interleukin-17/immunology , Intestines/immunology , Intestines/microbiology , Intestines/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , Th17 Cells/pathology
15.
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
16.
Mucosal Immunol ; 9(2): 352-63, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26242598

ABSTRACT

In inflammatory bowel diseases, a breakdown in host microbial interactions accompanies sustained activation of immune cells in the gut. Functional studies suggest a key role for interleukin-23 (IL-23) in orchestrating intestinal inflammation. IL-23 can be produced by various mononuclear phagocytes (MNPs) following acute microbial stimulation, but little is known about the key cellular sources of IL-23 that drive chronic intestinal inflammation. Here we have addressed this question using a physiological model of bacteria-driven colitis. By combining conditional gene ablation and gene expression profiling, we found that IL-23 production by CD11c(+) MNPs was essential to trigger intestinal immunopathology and identified MHCII(+) monocytes and macrophages as the major source of IL-23. Expression of IL-23 by monocytes was acquired during their differentiation in the intestine and correlated with the expression of major histocompatibility complex class II (MHCII) and CD64. In contrast, Batf3-dependent CD103(+) CD11b(-) dendritic cells were dispensable for bacteria-induced colitis in this model. These studies reinforce the pathogenic role of monocytes in dysregulated responses to intestinal bacteria and identify production of IL-23 as a key component of this response. Further understanding of the functional sources of IL-23 in diverse forms of intestinal inflammation may lead to novel therapeutic strategies aimed at interrupting IL-23-driven immune pathology.


Subject(s)
CD11c Antigen/immunology , Colitis/immunology , Helicobacter Infections/immunology , Interleukin-23/immunology , Intestinal Mucosa/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CD11c Antigen/genetics , Chronic Disease , Colitis/genetics , Colitis/microbiology , Colitis/pathology , Colon/immunology , Colon/pathology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gene Expression Profiling , Gene Expression Regulation , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter hepaticus/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Interleukin-23/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Transgenic , Monocytes/microbiology , Monocytes/pathology , Receptors, IgG/genetics , Receptors, IgG/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Repressor Proteins/genetics , Repressor Proteins/immunology , Signal Transduction
17.
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
18.
J Immunol ; 195(7): 3071-85, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26324769

ABSTRACT

It is clear that IL-10 plays an essential role in maintaining homeostasis in the gut in response to the microbiome. However, it is unknown whether IL-10 also facilitates immune homeostasis at distal sites. To address this question, we asked whether splenic immune populations were altered in IL-10-deficient (Il10(-/-)) mice in which differences in animal husbandry history were associated with susceptibility to spontaneous enterocolitis that is microbiome dependent. The susceptible mice exhibited a significant increase in splenic macrophages, neutrophils, and marginal zone (MZ) B cells that was inhibited by IL-10 signaling in myeloid, but not B cells. The increase in macrophages was due to increased proliferation that correlated with a subsequent enhancement in MZ B cell differentiation. Cohousing and antibiotic treatment studies suggested that the alteration in immune homeostasis in the spleen was microbiome dependent. The 16S rRNA sequencing revealed that susceptible mice harbored a different microbiome with a significant increase in the abundance of the bacterial genus Helicobacter. The introduction of Helicobacter hepaticus to the gut of nonsusceptible mice was sufficient to drive macrophage expansion and MZ B cell development. Given that myeloid cells and MZ B cells are part of the first line of defense against blood-borne pathogens, their increase following a breach in the gut epithelial barrier would be protective. Thus, IL-10 is an essential gatekeeper that maintains immune homeostasis at distal sites that can become functionally imbalanced upon the introduction of specific pathogenic bacteria to the intestinal track.


Subject(s)
B-Lymphocytes/immunology , Dysbiosis/microbiology , Gastrointestinal Microbiome/genetics , Helicobacter Infections/immunology , Helicobacter hepaticus/immunology , Interleukin-10/genetics , Animals , B-Lymphocytes/cytology , Base Sequence , Cell Count , Cell Differentiation/immunology , Cell Proliferation , DNA, Bacterial/genetics , Enterocolitis/immunology , Enterocolitis/microbiology , Helicobacter Infections/microbiology , Interleukin-10/immunology , Lymphocyte Activation/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Signal Transduction/immunology
19.
Oncotarget ; 6(11): 9387-96, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25831236

ABSTRACT

Recent studies suggest that gastrointestinal tract microbiota modulate cancer development in distant non-intestinal tissues. Here we tested mechanistic hypotheses using a targeted pathogenic gut microbial infection animal model with a predilection to breast cancer. FVB-Tg(C3-1-TAg)cJeg/JegJ female mice were infected by gastric gavage with Helicobacter hepaticus at three-months-of-age putting them at increased risk for mammary tumor development. Tumorigenesis was multifocal and characterized by extensive infiltrates of myeloperoxidase-positive neutrophils otherwise implicated in cancer progression in humans and animal models. To test whether neutrophils were important in etiopathogenesis in this bacteria-triggered model system, we next systemically depleted mice of neutrophils using thrice weekly intraperitoneal injections with anti-Ly-6G antibody. We found that antibody depletion entirely inhibited tumor development in this H. hepaticus-infected model. These data demonstrate that host neutrophil-associated immune responses to intestinal tract microbes significantly impact cancer progression in distal tissues such as mammary glands, and identify gut microbes as novel targets for extra-intestinal cancer therapy.


Subject(s)
Bacteria/immunology , Carcinogenesis/immunology , Intestines/microbiology , Mammary Neoplasms, Animal , Microbiota/physiology , Neutrophils/physiology , Animals , Female , Helicobacter Infections/immunology , Helicobacter hepaticus/immunology , Intestines/immunology , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/microbiology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic , Microbiota/immunology , Neutrophils/pathology
20.
Methods Mol Biol ; 1193: 199-211, 2014.
Article in English | MEDLINE | ID: mdl-25151008

ABSTRACT

Inflammatory bowel diseases (IBD) represent idiopathic chronic inflammatory disorders of the intestinal tract that are associated with aberrant immune responses against intestinal bacteria. Here, we describe two T cell-dependent models of experimental murine IBD. In the "T cell transfer" model, lymphopenic (scid or Rag (-/-) ) mice develop colitis upon adoptive transfer of naïve CD4(+) T cells. This model has also been extensively employed to identify mechanisms through which CD4(+)CD25(+) regulatory T cells suppress intestinal inflammation in vivo. We also describe a model of T cell-dependent IBD in immunocompetent mice, induced by infection with the intestinal bacterium Helicobacter hepaticus and concomitant treatment with a blocking αIL-10R mAb, which leads to the development of chronic inflammation of the caecum and colon (typhlocolitis). Both models reproduce many facets of human IBD pathology, including epithelial hyperplasia, goblet cell depletion, and leukocyte infiltration. These models provide reliable and tractable systems for the analyses of the induction and regulation of chronic inflammation in the gut.


Subject(s)
Disease Models, Animal , Inflammatory Bowel Diseases/immunology , Mice , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Animals , Disease Progression , Flow Cytometry , Helicobacter hepaticus/immunology , Homeodomain Proteins/genetics , Inflammatory Bowel Diseases/pathology , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Receptors, Interleukin-10/immunology , T-Lymphocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...