Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
Add more filters

Publication year range
1.
Cell ; 184(5): 1214-1231.e16, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33636133

ABSTRACT

Although enteric helminth infections modulate immunity to mucosal pathogens, their effects on systemic microbes remain less established. Here, we observe increased mortality in mice coinfected with the enteric helminth Heligmosomoides polygyrus bakeri (Hpb) and West Nile virus (WNV). This enhanced susceptibility is associated with altered gut morphology and transit, translocation of commensal bacteria, impaired WNV-specific T cell responses, and increased virus infection in the gastrointestinal tract and central nervous system. These outcomes were due to type 2 immune skewing, because coinfection in Stat6-/- mice rescues mortality, treatment of helminth-free WNV-infected mice with interleukin (IL)-4 mirrors coinfection, and IL-4 receptor signaling in intestinal epithelial cells mediates the susceptibility phenotypes. Moreover, tuft cell-deficient mice show improved outcomes with coinfection, whereas treatment of helminth-free mice with tuft cell-derived cytokine IL-25 or ligand succinate worsens WNV disease. Thus, helminth activation of tuft cell-IL-4-receptor circuits in the gut exacerbates infection and disease of a neurotropic flavivirus.


Subject(s)
Coinfection , Nematospiroides dubius/physiology , Signal Transduction , Strongylida Infections/pathology , West Nile virus/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Disease Susceptibility , Intestinal Mucosa/parasitology , Intestinal Mucosa/virology , Mice , Mice, Inbred C57BL , Neurons/parasitology , Neurons/virology , Receptors, Interleukin-4/metabolism , STAT6 Transcription Factor/genetics , Severity of Illness Index , Strongylida Infections/parasitology
2.
Cell ; 182(4): 901-918.e18, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32668198

ABSTRACT

Chikungunya virus (CHIKV), an emerging alphavirus, has infected millions of people. However, the factors modulating disease outcome remain poorly understood. Here, we show in germ-free mice or in oral antibiotic-treated conventionally housed mice with depleted intestinal microbiomes that greater CHIKV infection and spread occurs within 1 day of virus inoculation. Alteration of the microbiome alters TLR7-MyD88 signaling in plasmacytoid dendritic cells (pDCs) and blunts systemic production of type I interferon (IFN). Consequently, circulating monocytes express fewer IFN-stimulated genes and become permissive for CHIKV infection. Reconstitution with a single bacterial species, Clostridium scindens, or its derived metabolite, the secondary bile acid deoxycholic acid, can restore pDC- and MyD88-dependent type I IFN responses to restrict systemic CHIKV infection and transmission back to vector mosquitoes. Thus, symbiotic intestinal bacteria modulate antiviral immunity and levels of circulating alphaviruses within hours of infection through a bile acid-pDC-IFN signaling axis, which affects viremia, dissemination, and potentially transmission.


Subject(s)
Bile Acids and Salts/metabolism , Chikungunya Fever/pathology , Gastrointestinal Microbiome , Interferon Type I/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Chikungunya Fever/immunology , Chikungunya Fever/veterinary , Chikungunya virus/genetics , Chikungunya virus/isolation & purification , Clostridiales/physiology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/drug effects , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , RNA, Viral/blood , STAT1 Transcription Factor/deficiency , Signal Transduction , Toll-Like Receptor 7/metabolism
3.
Cell ; 179(5): 1144-1159.e15, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31708126

ABSTRACT

The colonic epithelium can undergo multiple rounds of damage and repair, often in response to excessive inflammation. The responsive stem cell that mediates this process is unclear, in part because of a lack of in vitro models that recapitulate key epithelial changes that occur in vivo during damage and repair. Here, we identify a Hopx+ colitis-associated regenerative stem cell (CARSC) population that functionally contributes to mucosal repair in mouse models of colitis. Hopx+ CARSCs, enriched for fetal-like markers, transiently arose from hypertrophic crypts known to facilitate regeneration. Importantly, we established a long-term, self-organizing two-dimensional (2D) epithelial monolayer system to model the regenerative properties and responses of Hopx+ CARSCs. This system can reenact the "homeostasis-injury-regeneration" cycles of epithelial alterations that occur in vivo. Using this system, we found that hypoxia and endoplasmic reticulum stress, insults commonly present in inflammatory bowel diseases, mediated the cyclic switch of cellular status in this process.


Subject(s)
Cell Culture Techniques/methods , Colon/pathology , Stem Cells/pathology , 3T3 Cells , Animals , Colitis/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Homeodomain Proteins/metabolism , Mice , Models, Biological , Oxygen/pharmacology , Regeneration/drug effects , Stem Cells/drug effects , Stress, Physiological/drug effects
4.
Cell ; 175(5): 1198-1212.e12, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30293866

ABSTRACT

Although chronic gastrointestinal dysmotility syndromes are a common worldwide health problem, underlying causes for these disorders are poorly understood. We show that flavivirus infection of enteric neurons leads to acute neuronal injury and cell death, inflammation, bowel dilation, and slowing of intestinal transit in mice. Flavivirus-primed CD8+ T cells promote these phenotypes, as their absence diminished enteric neuron injury and intestinal transit delays, and their adoptive transfer reestablished dysmotility after flavivirus infection. Remarkably, mice surviving acute flavivirus infection developed chronic gastrointestinal dysmotility that was exacerbated by immunization with an unrelated alphavirus vaccine or exposure to a non-infectious inflammatory stimulus. This model of chronic post-infectious gastrointestinal dysmotility in mice suggests that viral infections with tropism for enteric neurons and the ensuing immune response might contribute to the development of bowel motility disorders in humans. These results suggest an opportunity for unique approaches to diagnosis and therapy of gastrointestinal dysmotility syndromes.


Subject(s)
Flavivirus Infections/pathology , Flavivirus/pathogenicity , Gastrointestinal Motility , Intestines/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , Flavivirus/genetics , Flavivirus Infections/immunology , Flavivirus Infections/virology , Intestines/virology , Leukocytes/cytology , Leukocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Neurons/ultrastructure , RNA, Viral/isolation & purification , RNA, Viral/metabolism , Syndrome
5.
Nat Immunol ; 20(6): 687-700, 2019 06.
Article in English | MEDLINE | ID: mdl-31061528

ABSTRACT

Most tissue-resident macrophage populations develop during embryogenesis, self-renew in the steady state and expand during type 2 immunity. Whether shared mechanisms regulate the proliferation of macrophages in homeostasis and disease is unclear. Here we found that the transcription factor Bhlhe40 was required in a cell-intrinsic manner for the self-renewal and maintenance of large peritoneal macrophages (LPMs), but not that of other tissue-resident macrophages. Bhlhe40 was necessary for the proliferation, but not the polarization, of LPMs in response to the cytokine IL-4. During infection with the helminth Heligmosomoides polygyrus bakeri, Bhlhe40 was required for cell cycling of LPMs. Bhlhe40 repressed the expression of genes encoding the transcription factors c-Maf and Mafb and directly promoted expression of transcripts encoding cell cycle-related proteins to enable the proliferation of LPMs. In LPMs, Bhlhe40 bound to genomic sites co-bound by the macrophage lineage-determining factor PU.1 and to unique sites, including Maf and loci encoding cell-cycle-related proteins. Our findings demonstrate a tissue-specific control mechanism that regulates the proliferation of resident macrophages in homeostasis and type 2 immunity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Homeodomain Proteins/genetics , Homeostasis/genetics , Homeostasis/immunology , Immunity/genetics , Macrophages/immunology , Macrophages/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers , Cell Cycle/genetics , Cell Cycle/immunology , Cell Proliferation , Gene Expression Profiling , Gene Expression Regulation , Gene Knockout Techniques , Helicobacter Infections/genetics , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Homeodomain Proteins/metabolism , Immunophenotyping , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Transgenic , Monocytes/immunology , Monocytes/metabolism , Organ Specificity/genetics , Organ Specificity/immunology , Transcriptome
6.
Cell ; 165(7): 1708-1720, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27264604

ABSTRACT

In the mammalian intestine, crypts of Leiberkühn house intestinal epithelial stem/progenitor cells at their base. The mammalian intestine also harbors a diverse array of microbial metabolite compounds that potentially modulate stem/progenitor cell activity. Unbiased screening identified butyrate, a prominent bacterial metabolite, as a potent inhibitor of intestinal stem/progenitor proliferation at physiologic concentrations. During homeostasis, differentiated colonocytes metabolized butyrate likely preventing it from reaching proliferating epithelial stem/progenitor cells within the crypt. Exposure of stem/progenitor cells in vivo to butyrate through either mucosal injury or application to a naturally crypt-less host organism led to inhibition of proliferation and delayed wound repair. The mechanism of butyrate action depended on the transcription factor Foxo3. Our findings indicate that mammalian crypt architecture protects stem/progenitor cell proliferation in part through a metabolic barrier formed by differentiated colonocytes that consume butyrate and stimulate future studies on the interplay of host anatomy and microbiome metabolism.


Subject(s)
Bacteria/metabolism , Butyrates/metabolism , Colon/cytology , Colon/microbiology , Gastrointestinal Microbiome , Stem Cells/metabolism , Acyl-CoA Dehydrogenase/deficiency , Acyl-CoA Dehydrogenase/genetics , Animals , Cell Proliferation , Intestine, Small/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oxidation-Reduction , Pathogen-Associated Molecular Pattern Molecules/metabolism , Stem Cells/cytology , Zebrafish
7.
Cell ; 167(5): 1339-1353.e21, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863247

ABSTRACT

Despite the accepted health benefits of consuming dietary fiber, little is known about the mechanisms by which fiber deprivation impacts the gut microbiota and alters disease risk. Using a gnotobiotic mouse model, in which animals were colonized with a synthetic human gut microbiota composed of fully sequenced commensal bacteria, we elucidated the functional interactions between dietary fiber, the gut microbiota, and the colonic mucus barrier, which serves as a primary defense against enteric pathogens. We show that during chronic or intermittent dietary fiber deficiency, the gut microbiota resorts to host-secreted mucus glycoproteins as a nutrient source, leading to erosion of the colonic mucus barrier. Dietary fiber deprivation, together with a fiber-deprived, mucus-eroding microbiota, promotes greater epithelial access and lethal colitis by the mucosal pathogen, Citrobacter rodentium. Our work reveals intricate pathways linking diet, the gut microbiome, and intestinal barrier dysfunction, which could be exploited to improve health using dietary therapeutics.


Subject(s)
Dietary Fiber/administration & dosage , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Animals , Citrobacter rodentium/physiology , Colitis/microbiology , Colon/microbiology , Disease Susceptibility , Enterobacteriaceae Infections/microbiology , Escherichia coli , Female , Germ-Free Life , Humans , Male , Mice , Mucin-2/genetics
8.
Cell ; 160(3): 447-60, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25619688

ABSTRACT

Decreases in the diversity of enteric bacterial populations are observed in patients with Crohn's disease (CD) and ulcerative colitis (UC). Less is known about the virome in these diseases. We show that the enteric virome is abnormal in CD and UC patients. In-depth analysis of preparations enriched for free virions in the intestine revealed that CD and UC were associated with a significant expansion of Caudovirales bacteriophages. The viromes of CD and UC patients were disease and cohort specific. Importantly, it did not appear that expansion and diversification of the enteric virome was secondary to changes in bacterial populations. These data support a model in which changes in the virome may contribute to intestinal inflammation and bacterial dysbiosis. We conclude that the virome is a candidate for contributing to, or being a biomarker for, human inflammatory bowel disease and speculate that the enteric virome may play a role in other diseases.


Subject(s)
Caudovirales/isolation & purification , Colitis, Ulcerative/virology , Crohn Disease/virology , Dysbiosis/virology , Microviridae/isolation & purification , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Case-Control Studies , Caudovirales/genetics , Cohort Studies , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colitis, Ulcerative/therapy , Crohn Disease/microbiology , Crohn Disease/pathology , Crohn Disease/therapy , Dysbiosis/microbiology , Dysbiosis/pathology , Dysbiosis/therapy , Feces/microbiology , Feces/virology , Humans , Metagenome , Microviridae/genetics
9.
Cell ; 160(1-2): 269-84, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25594183

ABSTRACT

The stem cells that maintain and repair the postnatal skeleton remain undefined. One model suggests that perisinusoidal mesenchymal stem cells (MSCs) give rise to osteoblasts, chondrocytes, marrow stromal cells, and adipocytes, although the existence of these cells has not been proven through fate-mapping experiments. We demonstrate here that expression of the bone morphogenetic protein (BMP) antagonist gremlin 1 defines a population of osteochondroreticular (OCR) stem cells in the bone marrow. OCR stem cells self-renew and generate osteoblasts, chondrocytes, and reticular marrow stromal cells, but not adipocytes. OCR stem cells are concentrated within the metaphysis of long bones not in the perisinusoidal space and are needed for bone development, bone remodeling, and fracture repair. Grem1 expression also identifies intestinal reticular stem cells (iRSCs) that are cells of origin for the periepithelial intestinal mesenchymal sheath. Grem1 expression identifies distinct connective tissue stem cells in both the bone (OCR stem cells) and the intestine (iRSCs).


Subject(s)
Bone and Bones/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Intestine, Small/cytology , Mesenchymal Stem Cells/cytology , Animals , Cartilage/metabolism , Intestine, Small/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL
10.
Nat Immunol ; 16(11): 1124-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26414766

ABSTRACT

Subsets of innate lymphoid cells (ILCs) reside in the mucosa and regulate immune responses to external pathogens. While ILCs can be phenotypically classified into ILC1, ILC2 and ILC3 subsets, the transcriptional control of commitment to each ILC lineage is incompletely understood. Here we report that the transcription factor Runx3 was essential for the normal development of ILC1 and ILC3 cells but not of ILC2 cells. Runx3 controlled the survival of ILC1 cells but not of ILC3 cells. Runx3 was required for expression of the transcription factor RORγt and its downstream target, the transcription factor AHR, in ILC3 cells. The absence of Runx3 in ILCs exacerbated infection with Citrobacter rodentium. Therefore, our data establish Runx3 as a key transcription factor in the lineage-specific differentiation of ILC1 and ILC3 cells.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Animals , Antigens, Ly/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/immunology , Cell Lineage/immunology , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Core Binding Factor Alpha 3 Subunit/deficiency , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor beta Subunit/deficiency , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Enterobacteriaceae Infections/etiology , Enterobacteriaceae Infections/immunology , Interleukin-7 Receptor alpha Subunit/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Lymphocyte Subsets/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/deficiency , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
11.
Cell ; 151(2): 253-66, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23063120

ABSTRACT

Pathogenic simian immunodeficiency virus (SIV) infection is associated with enteropathy, which likely contributes to AIDS progression. To identify candidate etiologies for AIDS enteropathy, we used next-generation sequencing to define the enteric virome during SIV infection in nonhuman primates. Pathogenic, but not nonpathogenic, SIV infection was associated with significant expansion of the enteric virome. We identified at least 32 previously undescribed enteric viruses during pathogenic SIV infection and confirmed their presence by using viral culture and PCR testing. We detected unsuspected mucosal adenovirus infection associated with enteritis as well as parvovirus viremia in animals with advanced AIDS, indicating the pathogenic potential of SIV-associated expansion of the enteric virome. No association between pathogenic SIV infection and the family-level taxonomy of enteric bacteria was detected. Thus, enteric viral infections may contribute to AIDS enteropathy and disease progression. These findings underline the importance of metagenomic analysis of the virome for understanding AIDS pathogenesis.


Subject(s)
Caliciviridae/isolation & purification , Intestines/virology , Parvoviridae/isolation & purification , Picornaviridae/isolation & purification , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Animals , Caliciviridae/classification , Caliciviridae/genetics , Chlorocebus aethiops , Feces/microbiology , Feces/virology , Intestines/microbiology , Molecular Sequence Data , Parvoviridae/classification , Parvoviridae/genetics , Phylogeny , Picornaviridae/classification , Picornaviridae/genetics , Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/microbiology , Simian Immunodeficiency Virus/pathogenicity
13.
Nat Immunol ; 14(9): 937-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23913046

ABSTRACT

Defense against attaching-and-effacing bacteria requires the sequential generation of interleukin 23 (IL-23) and IL-22 to induce protective mucosal responses. Although CD4(+) and NKp46(+) innate lymphoid cells (ILCs) are the critical source of IL-22 during infection, the precise source of IL-23 is unclear. We used genetic techniques to deplete mice of specific subsets of classical dendritic cells (cDCs) and analyzed immunity to the attaching-and-effacing pathogen Citrobacter rodentium. We found that the signaling receptor Notch2 controlled the terminal stage of cDC differentiation. Notch2-dependent intestinal CD11b(+) cDCs were an obligate source of IL-23 required for survival after infection with C. rodentium, but CD103(+) cDCs dependent on the transcription factor Batf3 were not. Our results demonstrate a nonredundant function for CD11b(+) cDCs in the response to pathogens in vivo.


Subject(s)
Citrobacter rodentium/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Receptor, Notch2/metabolism , Animals , Antigens, CD/metabolism , CD11b Antigen/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Dendritic Cells/cytology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/mortality , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Interleukin-23/metabolism , Intestinal Mucosa/microbiology , Lectins, C-Type/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Transgenic , Minor Histocompatibility Antigens , Receptor, Notch2/deficiency , Receptors, Cell Surface/metabolism , Signal Transduction , Spleen/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Wound Healing/genetics , Wound Healing/immunology
14.
Cell ; 141(7): 1135-45, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20602997

ABSTRACT

It is unclear why disease occurs in only a small proportion of persons carrying common risk alleles of disease susceptibility genes. Here we demonstrate that an interaction between a specific virus infection and a mutation in the Crohn's disease susceptibility gene Atg16L1 induces intestinal pathologies in mice. This virus-plus-susceptibility gene interaction generated abnormalities in granule packaging and unique patterns of gene expression in Paneth cells. Further, the response to injury induced by the toxic substance dextran sodium sulfate was fundamentally altered to include pathologies resembling aspects of Crohn's disease. These pathologies triggered by virus-plus-susceptibility gene interaction were dependent on TNFalpha and IFNgamma and were prevented by treatment with broad spectrum antibiotics. Thus, we provide a specific example of how a virus-plus-susceptibility gene interaction can, in combination with additional environmental factors and commensal bacteria, determine the phenotype of hosts carrying common risk alleles for inflammatory disease.


Subject(s)
Carrier Proteins/genetics , Crohn Disease/genetics , Crohn Disease/virology , Genetic Predisposition to Disease , Ileum/pathology , Norovirus , Animals , Autophagy-Related Proteins , Crohn Disease/pathology , Gene Expression Profiling , Humans , Interferon-gamma/metabolism , Mice , Paneth Cells/metabolism , Paneth Cells/virology , Tumor Necrosis Factor-alpha/metabolism
15.
Nature ; 569(7758): 655-662, 2019 05.
Article in English | MEDLINE | ID: mdl-31142855

ABSTRACT

Inflammatory bowel diseases, which include Crohn's disease and ulcerative colitis, affect several million individuals worldwide. Crohn's disease and ulcerative colitis are complex diseases that are heterogeneous at the clinical, immunological, molecular, genetic, and microbial levels. Individual contributing factors have been the focus of extensive research. As part of the Integrative Human Microbiome Project (HMP2 or iHMP), we followed 132 subjects for one year each to generate integrated longitudinal molecular profiles of host and microbial activity during disease (up to 24 time points each; in total 2,965 stool, biopsy, and blood specimens). Here we present the results, which provide a comprehensive view of functional dysbiosis in the gut microbiome during inflammatory bowel disease activity. We demonstrate a characteristic increase in facultative anaerobes at the expense of obligate anaerobes, as well as molecular disruptions in microbial transcription (for example, among clostridia), metabolite pools (acylcarnitines, bile acids, and short-chain fatty acids), and levels of antibodies in host serum. Periods of disease activity were also marked by increases in temporal variability, with characteristic taxonomic, functional, and biochemical shifts. Finally, integrative analysis identified microbial, biochemical, and host factors central to this dysregulation. The study's infrastructure resources, results, and data, which are available through the Inflammatory Bowel Disease Multi'omics Database ( http://ibdmdb.org ), provide the most comprehensive description to date of host and microbial activities in inflammatory bowel diseases.


Subject(s)
Gastrointestinal Microbiome/genetics , Inflammatory Bowel Diseases/microbiology , Animals , Fungi/pathogenicity , Gastrointestinal Microbiome/immunology , Health , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/virology , Phylogeny , Species Specificity , Transcriptome , Viruses/pathogenicity
16.
Immunity ; 43(2): 216-8, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26287677

ABSTRACT

Microbially derived metabolites in the intestine regulate host immunity and impact disease pathophysiology in various organs. Sun et al. (2015) suggest a direct effect of microbial metabolites on pancreatic endocrine cells in regulating type 1 diabetes pathophysiology.


Subject(s)
Cathelicidins/metabolism , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Intestines/immunology , Microbiota/physiology , Pancreas/immunology , Animals , Female , Male
17.
J Immunol ; 209(4): 742-750, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35868637

ABSTRACT

The local microenvironment shapes macrophage differentiation in each tissue. We hypothesized that in the peritoneum, local factors in addition to retinoic acid can support GATA6-driven differentiation and function of peritoneal large cavity macrophages (LCMs). We found that soluble proteins produced by mesothelial cells lining the peritoneal cavity maintained GATA6 expression in cultured LCMs. Analysis of global gene expression of isolated mesothelial cells highlighted mesothelin (Msln) and its binding partner mucin 16 (Muc16) as candidate secreted ligands that potentially regulate GATA6 expression in peritoneal LCMs. Mice deficient for either of these molecules showed diminished GATA6 expression in peritoneal and pleural LCMs that was most prominent in aged mice. The more robust phenotype in older mice suggested that monocyte-derived macrophages were the target of Msln and Muc16. Cell transfer and bone marrow chimera experiments supported this hypothesis. We found that lethally irradiated Msln-/- and Muc16-/- mice reconstituted with wild-type bone marrow had lower levels of GATA6 expression in peritoneal and pleural LCMs. Similarly, during the resolution of zymosan-induced inflammation, repopulated peritoneal LCMs lacking expression of Msln or Muc16 expressed diminished GATA6. These data support a role for mesothelial cell-produced Msln and Muc16 in local macrophage differentiation within large cavity spaces such as the peritoneum. The effect appears to be most prominent on monocyte-derived macrophages that enter into this location as the host ages and also in response to infection.


Subject(s)
Macrophages, Peritoneal , Macrophages , Mice , Animals , Peritoneal Cavity , Peritoneum , Epithelium
18.
Gut ; 71(9): 1892-1908, 2022 09.
Article in English | MEDLINE | ID: mdl-35636923

ABSTRACT

We are entering an era of medicine where increasingly sophisticated data will be obtained from patients to determine proper diagnosis, predict outcomes and direct therapies. We predict that the most valuable data will be produced by systems that are highly dynamic in both time and space. Three-dimensional (3D) organoids are poised to be such a highly valuable system for a variety of gastrointestinal (GI) diseases. In the lab, organoids have emerged as powerful systems to model molecular and cellular processes orchestrating natural and pathophysiological human tissue formation in remarkable detail. Preclinical studies have impressively demonstrated that these organs-in-a-dish can be used to model immunological, neoplastic, metabolic or infectious GI disorders by taking advantage of patient-derived material. Technological breakthroughs now allow to study cellular communication and molecular mechanisms of interorgan cross-talk in health and disease including communication along for example, the gut-brain axis or gut-liver axis. Despite considerable success in culturing classical 3D organoids from various parts of the GI tract, some challenges remain to develop these systems to best help patients. Novel platforms such as organ-on-a-chip, engineered biomimetic systems including engineered organoids, micromanufacturing, bioprinting and enhanced rigour and reproducibility will open improved avenues for tissue engineering, as well as regenerative and personalised medicine. This review will highlight some of the established methods and also some exciting novel perspectives on organoids in the fields of gastroenterology. At present, this field is poised to move forward and impact many currently intractable GI diseases in the form of novel diagnostics and therapeutics.


Subject(s)
Bioprinting , Gastrointestinal Diseases , Gastrointestinal Diseases/diagnosis , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/therapy , Humans , Models, Theoretical , Organoids/metabolism , Reproducibility of Results
19.
Gut ; 71(7): 1289-1301, 2022 07.
Article in English | MEDLINE | ID: mdl-34261752

ABSTRACT

OBJECTIVE: Fibrosis is a common feature of Crohn's disease (CD) which can involve the mesenteric fat. However, the molecular signature of this process remains unclear. Our goal was to define the transcriptional signature of mesenteric fibrosis in CD subjects and to model mesenteric fibrosis in mice to improve our understanding of CD pathogenesis. DESIGN: We performed histological and transcriptional analysis of fibrosis in CD samples. We modelled a CD-like fibrosis phenotype by performing repeated colonic biopsies in mice and analysed the model by histology, type I collagen-targeted positron emission tomography (PET) and global gene expression. We generated a gene set list of essential features of mesenteric fibrosis and compared it to mucosal biopsy datasets from inflammatory bowel disease patients to identify a refined gene set that correlated with clinical outcomes. RESULTS: Mesenteric fibrosis in CD was interconnected to areas of fibrosis in all layers of the intestine, defined as penetrating fibrosis. We found a transcriptional signature of differentially expressed genes enriched in areas of the mesenteric fat of CD subjects with high levels of fibrosis. Mice subjected to repeated colonic biopsies showed penetrating fibrosis as shown by histology, PET imaging and transcriptional analysis. Finally, we composed a composite 24-gene set list that was linked to inflammatory fibroblasts and correlated with treatment response. CONCLUSION: We linked histopathological and molecular features of CD penetrating fibrosis to a mouse model of repeated biopsy injuries. This experimental system provides an innovative approach for functional investigations of underlying profibrotic mechanisms and therapeutic concepts in CD.


Subject(s)
Crohn Disease , Animals , Crohn Disease/complications , Crohn Disease/drug therapy , Crohn Disease/genetics , Fibrosis , Humans , Intestines/pathology , Mesentery/pathology , Mice , Tumor Necrosis Factor Inhibitors
20.
Gastroenterology ; 160(3): 809-822.e7, 2021 02.
Article in English | MEDLINE | ID: mdl-33160965

ABSTRACT

BACKGROUND AND AIMS: The host receptor for severe acute respiratory syndrome coronavirus 2, angiotensin-converting enzyme 2 (ACE2), is highly expressed in small bowel (SB). Our aim was to identify factors influencing intestinal ACE2 expression in Crohn's disease (CD), ulcerative colitis (UC), and non-inflammatory bowel disease (IBD) controls. METHODS: Using bulk RNA sequencing or microarray transcriptomics from tissue samples (4 SB and 2 colonic cohorts; n = 495; n = 387 UC; n = 94 non-IBD), we analyzed the relationship between ACE2 with demographics and disease activity and prognosis. We examined the outcome of anti-tumor necrosis factor and anti-interleukin-12/interleukin-23 treatment on SB and colonic ACE2 expression in 3 clinical trials. Univariate and multivariate regression models were fitted. RESULTS: ACE2 levels were consistently reduced in SB CD and elevated in colonic UC compared with non-IBD controls. Elevated SB ACE2 was also associated with demographic features (age and elevated body mass index) associated with poor coronavirus disease 2019 outcomes. Within CD, SB ACE2 was reduced in patients subsequently developing complicated disease. Within UC, colonic ACE2 was elevated in active disease and in patients subsequently requiring anti-tumor necrosis factor rescue therapy. SB and colonic ACE2 expression in active CD and UC were restored by anti-cytokine therapy, most notably in responders. CONCLUSIONS: Reduced SB but elevated colonic ACE2 levels in IBD are associated with inflammation and severe disease, but normalized after anti-cytokine therapy, suggesting compartmentalization of ACE2-related biology in SB and colonic inflammation. The restoration of ACE2 expression with anti-cytokine therapy might be important in the context of severe acute respiratory syndrome coronavirus 2 infection and potentially explain reports of reduced morbidity from coronavirus disease 2019 in IBD patients treated with anti-cytokines.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Intestines/drug effects , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2/genetics , Anti-Inflammatory Agents/adverse effects , COVID-19/enzymology , COVID-19/immunology , COVID-19/virology , Case-Control Studies , Child , Child, Preschool , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Crohn Disease/enzymology , Crohn Disease/genetics , Crohn Disease/immunology , Databases, Genetic , Female , Gene Expression Regulation, Enzymologic , Host-Pathogen Interactions , Humans , Intestines/enzymology , Intestines/immunology , Male , Middle Aged , North America , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Virus/metabolism , SARS-CoV-2/enzymology , SARS-CoV-2/immunology , Severity of Illness Index , Treatment Outcome , Tumor Necrosis Factor Inhibitors/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL