Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Annu Rev Immunol ; 36: 73-101, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29144836

ABSTRACT

The cellular degradative pathway of autophagy has a fundamental role in immunity. Here, we review the function of autophagy and autophagy proteins in inflammation. We discuss how the autophagy machinery controls the burden of infectious agents while simultaneously limiting inflammatory pathologies, which often involves processes that are distinct from conventional autophagy. Among the newly emerging processes we describe are LC3-associated phagocytosis and targeting by autophagy proteins, both of which require many of the same proteins that mediate conventional autophagy. We also discuss how autophagy contributes to differentiation of myeloid and lymphoid cell types, coordinates multicellular immunity, and facilitates memory responses. Together, these functions establish an intimate link between autophagy, mucosal immunity, and chronic inflammatory diseases. Finally, we offer our perspective on current challenges and barriers to translation.


Subject(s)
Autophagy , Disease Susceptibility , Inflammation/etiology , Animals , Biomarkers , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunomodulation , Inflammation/diagnosis , Inflammation/metabolism , Signal Transduction
2.
Nature ; 610(7932): 547-554, 2022 10.
Article in English | MEDLINE | ID: mdl-36198790

ABSTRACT

Loss of Paneth cells and their antimicrobial granules compromises the intestinal epithelial barrier and is associated with Crohn's disease, a major type of inflammatory bowel disease1-7. Non-classical lymphoid cells, broadly referred to as intraepithelial lymphocytes (IELs), intercalate the intestinal epithelium8,9. This anatomical position has implicated them as first-line defenders in resistance to infections, but their role in inflammatory disease pathogenesis requires clarification. The identification of mediators that coordinate crosstalk between specific IEL and epithelial subsets could provide insight into intestinal barrier mechanisms in health and disease. Here we show that the subset of IELs that express γ and δ T cell receptor subunits (γδ IELs) promotes the viability of Paneth cells deficient in the Crohn's disease susceptibility gene ATG16L1. Using an ex vivo lymphocyte-epithelium co-culture system, we identified apoptosis inhibitor 5 (API5) as a Paneth cell-protective factor secreted by γδ IELs. In the Atg16l1-mutant mouse model, viral infection induced a loss of Paneth cells and enhanced susceptibility to intestinal injury by inhibiting the secretion of API5 from γδ IELs. Therapeutic administration of recombinant API5 protected Paneth cells in vivo in mice and ex vivo in human organoids with the ATG16L1 risk allele. Thus, we identify API5 as a protective γδ IEL effector that masks genetic susceptibility to Paneth cell death.


Subject(s)
Apoptosis Regulatory Proteins , Crohn Disease , Genetic Predisposition to Disease , Intraepithelial Lymphocytes , Nuclear Proteins , Paneth Cells , Animals , Humans , Mice , Apoptosis Regulatory Proteins/metabolism , Cell Death , Crohn Disease/genetics , Crohn Disease/metabolism , Crohn Disease/pathology , Genetic Predisposition to Disease/genetics , Intestinal Mucosa/pathology , Nuclear Proteins/metabolism , Paneth Cells/pathology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Cell Survival , Organoids , Alleles
3.
Blood ; 135(26): 2388-2401, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32232483

ABSTRACT

A goal in precision medicine is to use patient-derived material to predict disease course and intervention outcomes. Here, we use mechanistic observations in a preclinical animal model to design an ex vivo platform that recreates genetic susceptibility to T-cell-mediated damage. Intestinal graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation. We found that intestinal GVHD in mice deficient in Atg16L1, an autophagy gene that is polymorphic in humans, is reversed by inhibiting necroptosis. We further show that cocultured allogeneic T cells kill Atg16L1-mutant intestinal organoids from mice, which was associated with an aberrant epithelial interferon signature. Using this information, we demonstrate that pharmacologically inhibiting necroptosis or interferon signaling protects human organoids derived from individuals harboring a common ATG16L1 variant from allogeneic T-cell attack. Our study provides a roadmap for applying findings in animal models to individualized therapy that targets affected tissues.


Subject(s)
Graft vs Host Disease/prevention & control , Intestinal Diseases/prevention & control , Organoids , T-Lymphocytes/immunology , Acrylamides/pharmacology , Animals , Autophagy , Autophagy-Related Proteins/deficiency , Autophagy-Related Proteins/genetics , Bone Marrow Transplantation/adverse effects , Coculture Techniques , Colon/abnormalities , Female , Genetic Predisposition to Disease , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Inflammatory Bowel Diseases/pathology , Intestinal Diseases/immunology , Intestinal Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Necroptosis/drug effects , Nitriles , Paneth Cells/pathology , Precision Medicine , Pyrazoles/pharmacology , Pyrimidines , Radiation Chimera , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Sulfonamides/pharmacology , T-Lymphocytes/transplantation
4.
J Clin Invest ; 130(4): 2111-2128, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31961824

ABSTRACT

Although Western diet and dysbiosis are the most prominent environmental factors associated with inflammatory bowel diseases (IBDs), the corresponding host factors and cellular mechanisms remain poorly defined. Here we report that the TSC1/mTOR pathway in the gut epithelium represents a metabolic and innate immune checkpoint for intestinal dysfunction and inflammation. mTOR hyperactivation triggered by Western diet or Tsc1 ablation led to epithelium necroptosis, barrier disruption, and predisposition to dextran sulfate sodium-induced colitis and inflammation-associated colon cancer. Mechanistically, our results uncovered a critical role for TSC1/mTOR in restraining the expression and activation of RIPK3 in the gut epithelium through TRIM11-mediated ubiquitination and autophagy-dependent degradation. Notably, microbiota depletion by antibiotics or gnotobiotics attenuated RIPK3 expression and activation, thereby alleviating epithelial necroptosis and colitis driven by mTOR hyperactivation. mTOR primarily impinged on RIPK3 to potentiate necroptosis induced by TNF and by microbial pathogen-associated molecular patterns (PAMPs), and hyperactive mTOR and aberrant necroptosis were intertwined in human IBDs. Together, our data reveal a previously unsuspected link between the Western diet, microbiota, and necroptosis and identify the mTOR/RIPK3/necroptosis axis as a driving force for intestinal inflammation and cancer.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein/metabolism , Animals , Inflammation , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Mice , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis Complex 1 Protein/genetics
5.
Nat Microbiol ; 4(10): 1737-1749, 2019 10.
Article in English | MEDLINE | ID: mdl-31182797

ABSTRACT

Products derived from bacterial members of the gut microbiota evoke immune signalling pathways of the host that promote immunity and barrier function in the intestine. How immune reactions to enteric viruses support intestinal homeostasis is unknown. We recently demonstrated that infection by murine norovirus (MNV) reverses intestinal abnormalities following depletion of bacteria, indicating that an intestinal animal virus can provide cues to the host that are typically attributed to the microbiota. Here, we elucidate mechanisms by which MNV evokes protective responses from the host. We identify an important role for the viral protein NS1/2 in establishing local replication and a type I interferon (IFN-I) response in the colon. We further show that IFN-I acts on intestinal epithelial cells to increase the proportion of CCR2-dependent macrophages and interleukin (IL)-22-producing innate lymphoid cells, which in turn promote pSTAT3 signalling in intestinal epithelial cells and protection from intestinal injury. In addition, we demonstrate that MNV provides a striking IL-22-dependent protection against early-life lethal infection by Citrobacter rodentium. These findings demonstrate novel ways in which a viral member of the microbiota fortifies the intestinal barrier during chemical injury and infectious challenges.


Subject(s)
Gastrointestinal Microbiome/immunology , Interferon Type I/metabolism , Interleukins/metabolism , Intestines/immunology , Intestines/virology , Animals , Anti-Bacterial Agents/toxicity , Cell Proliferation , Citrobacter rodentium/physiology , Colon/cytology , Colon/immunology , Colon/metabolism , Colon/virology , Dextran Sulfate/toxicity , Enterobacteriaceae Infections/prevention & control , Interleukins/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/drug effects , Lymphocytes/cytology , Lymphocytes/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Norovirus/immunology , Norovirus/physiology , Signal Transduction/genetics , Specific Pathogen-Free Organisms , Viral Nonstructural Proteins/genetics , Virus Replication , Interleukin-22
6.
J Exp Med ; 214(12): 3687-3705, 2017 Dec 04.
Article in English | MEDLINE | ID: mdl-29089374

ABSTRACT

A variant of the autophagy gene ATG16L1 is associated with Crohn's disease, an inflammatory bowel disease (IBD), and poor survival in allogeneic hematopoietic stem cell transplant recipients. We demonstrate that ATG16L1 in the intestinal epithelium is essential for preventing loss of Paneth cells and exaggerated cell death in animal models of virally triggered IBD and allogeneic hematopoietic stem cell transplantation. Intestinal organoids lacking ATG16L1 reproduced this loss in Paneth cells and displayed TNFα-mediated necroptosis, a form of programmed necrosis. This cytoprotective function of ATG16L1 was associated with the role of autophagy in promoting mitochondrial homeostasis. Finally, therapeutic blockade of necroptosis through TNFα or RIPK1 inhibition ameliorated disease in the virally triggered IBD model. These findings indicate that, in contrast to tumor cells in which autophagy promotes caspase-independent cell death, ATG16L1 maintains the intestinal barrier by inhibiting necroptosis in the epithelium.


Subject(s)
Apoptosis , Autophagy , Carrier Proteins/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Animals , Autophagy-Related Proteins , Caliciviridae Infections/pathology , Caliciviridae Infections/virology , Cell Survival , Cytoprotection , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Deletion , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Hematopoietic Stem Cell Transplantation , Homeostasis , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/ultrastructure , Mutation/genetics , Necrosis , Norovirus/physiology , Organoids/pathology , Paneth Cells/metabolism , Paneth Cells/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL