Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Cell ; 184(19): 5015-5030.e16, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34407392

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) regulate immunity and inflammation, yet their role in cancer remains elusive. Here, we identify that colorectal cancer (CRC) manifests with altered ILC3s that are characterized by reduced frequencies, increased plasticity, and an imbalance with T cells. We evaluated the consequences of these changes in mice and determined that a dialog between ILC3s and T cells via major histocompatibility complex class II (MHCII) is necessary to support colonization with microbiota that subsequently induce type-1 immunity in the intestine and tumor microenvironment. As a result, mice lacking ILC3-specific MHCII develop invasive CRC and resistance to anti-PD-1 immunotherapy. Finally, humans with dysregulated intestinal ILC3s harbor microbiota that fail to induce type-1 immunity and immunotherapy responsiveness when transferred to mice. Collectively, these data define a protective role for ILC3s in cancer and indicate that their inherent disruption in CRC drives dysfunctional adaptive immunity, tumor progression, and immunotherapy resistance.


Subject(s)
Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Disease Progression , Immunity, Innate , Immunotherapy , Lymphocytes/immunology , Animals , Cell Communication/drug effects , Cell Plasticity/drug effects , Colonic Neoplasms/microbiology , Feces/microbiology , Histocompatibility Antigens Class II/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunity, Innate/drug effects , Inflammation/immunology , Inflammation/pathology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Intestines/pathology , Lymphocytes/drug effects , Mice, Inbred C57BL , Microbiota/drug effects , Neoplasm Invasiveness , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tissue Donors
2.
Nat Immunol ; 23(2): 251-261, 2022 02.
Article in English | MEDLINE | ID: mdl-35102343

ABSTRACT

Tumor necrosis factor (TNF) drives chronic inflammation and cell death in the intestine, and blocking TNF is a therapeutic approach in inflammatory bowel disease (IBD). Despite this knowledge, the pathways that protect the intestine from TNF are incompletely understood. Here we demonstrate that group 3 innate lymphoid cells (ILC3s) protect the intestinal epithelium from TNF-induced cell death. This occurs independent of interleukin-22 (IL-22), and we identify that ILC3s are a dominant source of heparin-binding epidermal growth factor-like growth factor (HB-EGF). ILC3s produce HB-EGF in response to prostaglandin E2 (PGE2) and engagement of the EP2 receptor. Mice lacking ILC3-derived HB-EGF exhibit increased susceptibility to TNF-mediated epithelial cell death and experimental intestinal inflammation. Finally, human ILC3s produce HB-EGF and are reduced from the inflamed intestine. These results define an essential role for ILC3-derived HB-EGF in protecting the intestine from TNF and indicate that disruption of this pathway contributes to IBD.


Subject(s)
Heparin-binding EGF-like Growth Factor/immunology , Immunity, Innate/immunology , Inflammation/immunology , Intestines/immunology , Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Epithelial Cells/immunology , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology
3.
Nature ; 630(8018): 976-983, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867048

ABSTRACT

Interleukin (IL-)23 is a major mediator and therapeutic target in chronic inflammatory diseases that also elicits tissue protection in the intestine at homeostasis or following acute infection1-4. However, the mechanisms that shape these beneficial versus pathological outcomes remain poorly understood. To address this gap in knowledge, we performed single-cell RNA sequencing on all IL-23 receptor-expressing cells in the intestine and their acute response to IL-23, revealing a dominance of T cells and group 3 innate lymphoid cells (ILC3s). Unexpectedly, we identified potent upregulation of the immunoregulatory checkpoint molecule cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) on ILC3s. This pathway was activated by gut microbes and IL-23 in a FOXO1- and STAT3-dependent manner. Mice lacking CTLA-4 on ILC3s exhibited reduced regulatory T cells, elevated inflammatory T cells and more-severe intestinal inflammation. IL-23 induction of CTLA-4+ ILC3s was necessary and sufficient to reduce co-stimulatory molecules and increase PD-L1 bioavailability on intestinal myeloid cells. Finally, human ILC3s upregulated CTLA-4 in response to IL-23 or gut inflammation and correlated with immunoregulation in inflammatory bowel disease. These results reveal ILC3-intrinsic CTLA-4 as an essential checkpoint that restrains the pathological outcomes of IL-23, suggesting that disruption of these lymphocytes, which occurs in inflammatory bowel disease5-7, contributes to chronic inflammation.


Subject(s)
Immunity, Innate , Inflammation , Interleukin-23 , Lymphocytes , Animals , Female , Humans , Male , Mice , CTLA-4 Antigen/metabolism , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Gastrointestinal Microbiome , Inflammation/immunology , Inflammation/pathology , Inflammation/metabolism , Interleukin-23/immunology , Intestines/immunology , Intestines/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred C57BL , Myeloid Cells/metabolism , Single-Cell Gene Expression Analysis , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
4.
Nat Immunol ; 18(8): 851-860, 2017 Jul 19.
Article in English | MEDLINE | ID: mdl-28722709

ABSTRACT

The study of the intestinal microbiota has begun to shift from cataloging individual members of the commensal community to understanding their contributions to the physiology of the host organism in health and disease. Here, we review the effects of the microbiome on innate and adaptive immunological players from epithelial cells and antigen-presenting cells to innate lymphoid cells and regulatory T cells. We discuss recent studies that have identified diverse microbiota-derived bioactive molecules and their effects on inflammation within the intestine and distally at sites as anatomically remote as the brain. Finally, we highlight new insights into how the microbiome influences the host response to infection, vaccination and cancer, as well as susceptibility to autoimmune and neurodegenerative disorders.


Subject(s)
Gastrointestinal Microbiome/immunology , Infections/immunology , Inflammation/immunology , Neoplasms/immunology , Adaptive Immunity/immunology , Antigen-Presenting Cells/immunology , Autoimmune Diseases/immunology , Humans , Immunity, Innate/immunology , Immunity, Mucosal/immunology , Lymphocytes/immunology , Neurodegenerative Diseases/immunology , Symbiosis , T-Lymphocytes, Regulatory/immunology , Vaccination
5.
Immunity ; 52(2): 207-209, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075721

ABSTRACT

Innate lymphoid cells (ILCs) are tissue-resident lymphocytes that promote immunity to pathogens at mucosal barriers, but the mechanisms regulating their development within tissues remain poorly understood. In this issue of Immunity, Oherle et al. identify a niche in the neonatal airway where stromal cell-derived insulin-like growth factor 1 (IGF1) supports the proliferation of ILC precursors and protects from infection.


Subject(s)
Immunity, Innate , Lymphocytes , Humans , Infant, Newborn , Insulin-Like Growth Factor I , Lung
6.
Nature ; 609(7925): 159-165, 2022 09.
Article in English | MEDLINE | ID: mdl-35831503

ABSTRACT

RORγt is a lineage-specifying transcription factor that is expressed by immune cells that are enriched in the gastrointestinal tract and promote immunity, inflammation and tissue homeostasis1-15. However, fundamental questions remain with regard to the cellular heterogeneity among these cell types, the mechanisms that control protective versus inflammatory properties and their functional redundancy. Here we define all RORγt+ immune cells in the intestine at single-cell resolution and identify a subset of group 3 innate lymphoid cells (ILC3s) that expresses ZBTB46, a transcription factor specifying conventional dendritic cells16-20. ZBTB46 is robustly expressed by CCR6+ lymphoid-tissue-inducer-like ILC3s that are developmentally and phenotypically distinct from conventional dendritic cells, and its expression is imprinted by RORγt, fine-tuned by microbiota-derived signals and increased by pro-inflammatory cytokines. ZBTB46 restrains the inflammatory properties of ILC3s, including the OX40L-dependent expansion of T helper 17 cells and the exacerbated intestinal inflammation that occurs after enteric infection. Finally, ZBTB46+ ILC3s are a major source of IL-22, and selective depletion of this population renders mice susceptible to enteric infection and associated intestinal inflammation. These results show that ZBTB46 is a transcription factor that is shared between conventional dendritic cells and ILC3s, and identify a cell-intrinsic function for ZBTB46 in restraining the pro-inflammatory properties of ILC3s and a non-redundant role for ZBTB46+ ILC3s in orchestrating intestinal health.


Subject(s)
Immunity, Innate , Intestines , Lymphocytes , Nuclear Receptor Subfamily 1, Group F, Member 3 , Transcription Factors , Animals , Inflammation/immunology , Inflammation/pathology , Interleukins , Intestines/cytology , Intestines/immunology , Intestines/pathology , Lymphocytes/cytology , Lymphocytes/immunology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , OX40 Ligand/metabolism , Receptors, CCR6/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Transcription Factors/metabolism , Interleukin-22
7.
Nature ; 610(7933): 744-751, 2022 10.
Article in English | MEDLINE | ID: mdl-36071169

ABSTRACT

Microbial colonization of the mammalian intestine elicits inflammatory or tolerogenic T cell responses, but the mechanisms controlling these distinct outcomes remain poorly understood, and accumulating evidence indicates that aberrant immunity to intestinal microbiota is causally associated with infectious, inflammatory and malignant diseases1-8. Here we define a critical pathway controlling the fate of inflammatory versus tolerogenic T cells that respond to the microbiota and express the transcription factor RORγt. We profiled all RORγt+ immune cells at single-cell resolution from the intestine-draining lymph nodes of mice and reveal a dominant presence of T regulatory (Treg) cells and lymphoid tissue inducer-like group 3 innate lymphoid cells (ILC3s), which co-localize at interfollicular regions. These ILC3s are distinct from extrathymic AIRE-expressing cells, abundantly express major histocompatibility complex class II, and are necessary and sufficient to promote microbiota-specific RORγt+ Treg cells and prevent their expansion as inflammatory T helper 17 cells. This occurs through ILC3-mediated antigen presentation, αV integrin and competition for interleukin-2. Finally, single-cell analyses suggest that interactions between ILC3s and RORγt+ Treg cells are impaired in inflammatory bowel disease. Our results define a paradigm whereby ILC3s select for antigen-specific RORγt+ Treg cells, and against T helper 17 cells, to establish immune tolerance to the microbiota and intestinal health.


Subject(s)
Immune Tolerance , Intestines , Lymphocytes , Microbiota , T-Lymphocytes, Regulatory , Animals , Immunity, Innate , Integrin alphaV/metabolism , Interleukin-2/immunology , Intestines/immunology , Intestines/microbiology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocytes/immunology , Microbiota/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Single-Cell Analysis , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transcription Factors/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology
8.
Nature ; 600(7890): 707-712, 2021 12.
Article in English | MEDLINE | ID: mdl-34853467

ABSTRACT

Pro-inflammatory T cells in the central nervous system (CNS) are causally associated with multiple demyelinating and neurodegenerative diseases1-6, but the pathways that control these responses remain unclear. Here we define a population of inflammatory group 3 innate lymphoid cells (ILC3s) that infiltrate the CNS in a mouse model of multiple sclerosis. These ILC3s are derived from the circulation, localize in proximity to infiltrating T cells in the CNS, function as antigen-presenting cells that restimulate myelin-specific T cells, and are increased in individuals with multiple sclerosis. Notably, antigen presentation by inflammatory ILC3s is required to promote T cell responses in the CNS and the development of multiple-sclerosis-like disease in mouse models. By contrast, conventional and tissue-resident ILC3s in the periphery do not appear to contribute to disease induction, but instead limit autoimmune T cell responses and prevent multiple-sclerosis-like disease when experimentally targeted to present myelin antigen. Collectively, our data define a population of inflammatory ILC3s that is essential for directly promoting T-cell-dependent neuroinflammation in the CNS and reveal the potential of harnessing peripheral tissue-resident ILC3s for the prevention of autoimmune disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Antigen-Presenting Cells , Antigens/metabolism , Immunity, Innate , Lymphocytes , Mice , Neuroinflammatory Diseases , Sclerosis/metabolism
9.
Immunity ; 44(3): 634-646, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26982365

ABSTRACT

Physical separation between the mammalian immune system and commensal bacteria is necessary to limit chronic inflammation. However, selective species of commensal bacteria can reside within intestinal lymphoid tissues of healthy mammals. Here, we demonstrate that lymphoid-tissue-resident commensal bacteria (LRC) colonized murine dendritic cells and modulated their cytokine production. In germ-free and antibiotic-treated mice, LRCs colonized intestinal lymphoid tissues and induced multiple members of the IL-10 cytokine family, including dendritic-cell-derived IL-10 and group 3 innate lymphoid cell (ILC3)-derived IL-22. Notably, IL-10 limited the development of pro-inflammatory Th17 cell responses, and IL-22 production enhanced LRC colonization in the steady state. Furthermore, LRC colonization protected mice from lethal intestinal damage in an IL-10-IL-10R-dependent manner. Collectively, our data reveal a unique host-commensal-bacteria dialog whereby selective subsets of commensal bacteria interact with dendritic cells to facilitate tissue-specific responses that are mutually beneficial for both the host and the microbe.


Subject(s)
Bordetella Infections/immunology , Bordetella/immunology , Dendritic Cells/immunology , Interleukin-10/metabolism , Intestines/immunology , Lymphoid Tissue/immunology , Th17 Cells/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/microbiology , Interleukin-10/genetics , Interleukins/genetics , Interleukins/metabolism , Intestines/microbiology , Lymphoid Tissue/microbiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Receptors, Interleukin-10/genetics , Receptors, Interleukin-10/metabolism , Symbiosis/genetics , Th17 Cells/microbiology , Interleukin-22
10.
Nature ; 568(7752): 405-409, 2019 04.
Article in English | MEDLINE | ID: mdl-30944470

ABSTRACT

Interleukin (IL)-2 is a pleiotropic cytokine that is necessary to prevent chronic inflammation in the gastrointestinal tract1-4. The protective effects of IL-2 involve the generation, maintenance and function of regulatory T (Treg) cells4-8, and the use of low doses of IL-2 has emerged as a potential therapeutic strategy for patients with inflammatory bowel disease9. However, the cellular and molecular pathways that control the production of IL-2 in the context of intestinal health are undefined. Here we show, in a mouse model, that IL-2 is acutely required to maintain Treg cells and immunological homeostasis throughout the gastrointestinal tract. Notably, lineage-specific deletion of IL-2 in T cells did not reduce Treg cells in the small intestine. Unbiased analyses revealed that, in the small intestine, group-3 innate lymphoid cells (ILC3s) are the dominant cellular source of IL-2, which is induced selectively by IL-1ß. Macrophages in the small intestine produce IL-1ß, and activation of this pathway involves MYD88- and NOD2-dependent sensing of the microbiota. Our loss-of-function studies show that ILC3-derived IL-2 is essential for maintaining Treg cells, immunological homeostasis and oral tolerance to dietary antigens in the small intestine. Furthermore, production of IL-2 by ILC3s was significantly reduced in the small intestine of patients with Crohn's disease, and this correlated with lower frequencies of Treg cells. Our results reveal a previously unappreciated pathway in which a microbiota- and IL-1ß-dependent axis promotes the production of IL-2 by ILC3s to orchestrate immune regulation in the intestine.


Subject(s)
Immunity, Innate/immunology , Interleukin-2/immunology , Intestines/cytology , Intestines/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/administration & dosage , Antigens/immunology , Crohn Disease/immunology , Crohn Disease/metabolism , Crohn Disease/pathology , Female , Gastrointestinal Microbiome/immunology , Homeostasis/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-2/deficiency , Interleukin-2/metabolism , Intestine, Small/cytology , Intestine, Small/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , T-Lymphocytes, Regulatory/classification , T-Lymphocytes, Regulatory/metabolism
12.
Nat Immunol ; 12(5): 383-90, 2011 May.
Article in English | MEDLINE | ID: mdl-21502992

ABSTRACT

The maintenance of barrier function at exposed surfaces of the mammalian body is essential for limiting exposure to environmental stimuli, preventing systemic dissemination of commensal and pathogenic microbes and retaining normal homeostasis of the entire body. Indeed, dysregulated barrier function is associated with many infectious and inflammatory diseases, including psoriasis, influenza, inflammatory bowel disease and human immunodeficiency virus, which collectively afflict millions of people worldwide. Studies have shown that interleukin 22 (IL-22) is expressed at barrier surfaces and that its expression is dysregulated in certain human diseases, which suggests a critical role in the maintenance of normal barrier homeostasis. Consistent with that, studies of mouse model systems have identified a critical role for signaling by IL-22 through its receptor (IL-22R) in the promotion of antimicrobial immunity, inflammation and tissue repair at barrier surfaces. In this review we will discuss how the expression of IL-22 and IL-22R is regulated, the functions of the IL-22-IL-22R pathway in regulating immunity, inflammation and tissue homeostasis, and the therapeutic potential of targeting this pathway in human disease.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Inflammation/immunology , Interleukins/immunology , Animals , Humans , Intestines/immunology , Mice , Models, Animal , Receptors, Interleukin/immunology , Respiratory System/immunology , Signal Transduction/immunology , Skin/immunology , Interleukin-22
13.
Nat Immunol ; 12(11): 1045-54, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21946417

ABSTRACT

Innate lymphoid cells (ILCs), a heterogeneous cell population, are critical in orchestrating immunity and inflammation in the intestine, but whether ILCs influence immune responses or tissue homeostasis at other mucosal sites remains poorly characterized. Here we identify a population of lung-resident ILCs in mice and humans that expressed the alloantigen Thy-1 (CD90), interleukin 2 (IL-2) receptor a-chain (CD25), IL-7 receptor a-chain (CD127) and the IL-33 receptor subunit T1-ST2. Notably, mouse ILCs accumulated in the lung after infection with influenza virus, and depletion of ILCs resulted in loss of airway epithelial integrity, diminished lung function and impaired airway remodeling. These defects were restored by administration of the lung ILC product amphiregulin. Collectively, our results demonstrate a critical role for lung ILCs in restoring airway epithelial integrity and tissue homeostasis after infection with influenza virus.


Subject(s)
Homeostasis , Immunity, Innate , Influenza, Human/immunology , Lung/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Respiratory Mucosa/metabolism , Airway Remodeling/drug effects , Airway Remodeling/immunology , Amphiregulin , Animals , Antigens, CD/biosynthesis , Cells, Cultured , EGF Family of Proteins , Glycoproteins/pharmacology , Homeostasis/immunology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Interleukin-33 , Interleukins/metabolism , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred C57BL , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Wound Healing
14.
Trends Immunol ; 41(8): 721-733, 2020 08.
Article in English | MEDLINE | ID: mdl-32646594

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) have emerged as master regulators of intestinal health and tissue homeostasis in mammals. Through a diverse array of cytokines and cellular interactions, ILC3s crucially orchestrate lymphoid organogenesis, promote tissue protection or regeneration, facilitate antimicrobial responses, and directly regulate adaptive immunity. Further, translational studies have found that ILC3 responses are altered in the intestine of defined patient populations with chronic infectious, inflammatory, or metabolic diseases. Therefore, it is essential to broadly understand the signals that activate, suppress, or fine-tune ILC3s in the gut. Here, we discuss recent exciting advances in this field, integrate them into our current understanding of ILC3 biology, and highlight fundamental gaps in knowledge that require additional investigation.


Subject(s)
Gastrointestinal Microbiome , Immunity, Innate , Lymphocytes , Adaptive Immunity/immunology , Animals , Gastrointestinal Microbiome/immunology , Homeostasis , Humans , Immunity, Innate/immunology , Lymphocytes/cytology , Lymphocytes/immunology
15.
Immunity ; 39(2): 386-99, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23954130

ABSTRACT

Aryl hydrocarbon receptor (Ahr) is crucial for the maintenance and function of group 3 innate lymphoid cells (ILCs), which are important in gut immunity. Because Ahr promotes T helper 17 (Th17) cell differentiation in vitro, it is reasonable to expect that Ahr would enhance Th17 cells in vivo. Instead, we show that Ahr deficiency caused increased intestinal Th17 cells, raising the possibility that group 3 ILCs could negatively regulate Th17 cells. Reduced innate interleukin-22 (IL-22) in Ahr-deficient mice allowed expansion of commensal segmented filamentous bacteria (SFB), known to promote Th17 cells. Compared to Rorc(+/+)Ahr(-/-) mice, Rorc(gfp/+)Ahr(-/-) mice had further reduced group 3 ILCs and were prone to spontaneous colitis with increased SFB and Th17 cells. Innate expression of Ahr played a protective role in T-cell-mediated experimental colitis by suppressing pathogenic Th17 cells. Our data reveal an intricate balance between ILCs and Th17 cells regulated by Ahr and commensal flora.


Subject(s)
Colitis/immunology , Interleukins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Th17 Cells/immunology , Animals , Cell Differentiation/immunology , Interleukin-17/metabolism , Interleukins/biosynthesis , Intestines/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Th17 Cells/metabolism , Interleukin-22
16.
Immunity ; 38(4): 694-704, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601684

ABSTRACT

Group 2 innate lymphoid cells (ILC2) are innate lymphocytes that confer protective type 2 immunity during helminth infection and are also involved in allergic airway inflammation. Here we report that ILC2 development required T cell factor 1 (TCF-1, the product of the Tcf7 gene), a transcription factor also implicated in T cell lineage specification. Tcf7(-/-) mice lack ILC2, and were unable to mount ILC2-mediated innate type 2 immune responses. Forced expression of TCF-1 in bone marrow progenitors partially bypassed the requirement for Notch signaling in the generation of ILC2 in vivo. TCF-1 acted through both GATA-3-dependent and GATA-3-independent pathways to promote the generation of ILC2. These results are reminiscent of the critical roles of TCF-1 in early T cell development. Hence, transcription factors that underlie early steps of T cell development are also implicated in the development of innate lymphoid cells.


Subject(s)
Asthma/immunology , Bone Marrow Cells/immunology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Lymphocytes/immunology , Nippostrongylus/immunology , Strongylida Infections/immunology , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Hepatocyte Nuclear Factor 1-alpha/genetics , Immunity, Innate , Lymphoid Progenitor Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Transgenes/genetics
17.
Adv Exp Med Biol ; 1365: 113-134, 2022.
Article in English | MEDLINE | ID: mdl-35567744

ABSTRACT

Mucosal barrier surfaces of the mammalian body are frequent sites of pathogen colonization or entry and are also densely colonized with trillions of normally beneficial microbes, termed the microbiota. Therefore, it is paramount that the host immune system recognizes these microbes and is capable of differentiating between them. To this end, a multitude of mechanisms have evolved to carefully balance the need for immune activation in the face of infections while maintaining an appropriate level of tolerance to protect both the host and the beneficial microbes from hyperactivation. These mechanisms include the deployment of an emerging class of tissue-resident innate immune cells, innate lymphoid cells (ILCs), that are enriched at mucosal barriers such as the lungs and intestines, and are critical mediators of tissue homeostasis, tolerance, repair, and innate immunity. Recent findings have provided insight into the regulation of these cells and their interactions, not only with microbes, both commensal and foreign, but also with other systems of the body to prevent disease and promote tissue health. Here, we discuss recent findings in the regulation and function of ILCs, including a focus on their interactions with bodily systems, such as the nervous system, and how these interactions affect their functionality in states of health, infection, and disease.


Subject(s)
Immunity, Mucosal , Microbiota , Animals , Immunity, Innate , Lymphocytes , Mammals , Mucous Membrane
18.
Immunol Rev ; 286(1): 137-147, 2018 11.
Article in English | MEDLINE | ID: mdl-30294971

ABSTRACT

Innate and adaptive lymphocytes employ diverse effector programs that provide optimal immunity to pathogens and orchestrate tissue homeostasis, or conversely can become dysregulated to drive progression of chronic inflammatory diseases. Emerging evidence suggests that CD4+ T helper cell subsets and their innate counterparts, the innate lymphoid cell family, accomplish these complex biological roles by selectively programming their cellular metabolism in order to instruct distinct modules of lymphocyte differentiation, proliferation, and cytokine production. Further, these metabolic pathways are significantly influenced by tissue microenvironments and disease states. Here, we summarize our current knowledge on how cell-intrinsic metabolic factors modulate the context-dependent bioenergetic pathways that govern innate and adaptive lymphocytes. Further, we propose that a greater understanding of these pathways may lead to the identification of unique features in each population and provoke the development of novel therapeutic strategies to modulate lymphocytes in health and disease.


Subject(s)
Adaptive Immunity , Immunity, Innate , Lymphocytes/metabolism , Animals , Cell Differentiation , Cellular Microenvironment , Energy Metabolism , Humans , Immunomodulation , Lymphocyte Activation , Lymphocytes/immunology
19.
Immunity ; 37(4): 601-10, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-23084357

ABSTRACT

The mammalian intestine harbors trillions of beneficial commensal bacteria that are essential for the development of the immune system and for maintenance of physiologic processes in multiple organs. However, numerous chronic infectious, inflammatory, and metabolic diseases in humans have been associated with alterations in the composition or localization of commensal bacteria that result in dysregulated host-commensal bacteria relationships. The mammalian immune system plays an essential role in regulating the acquisition, composition, and localization of commensal bacteria in the intestine. Emerging research has implicated innate lymphoid cells (ILCs) as a critical immune cell population that orchestrates some of these host-commensal bacteria relationships that can impact immunity, inflammation, and tissue homeostasis in the intestine. This review will discuss reciprocal interactions between intestinal commensal bacteria and ILCs in the context of health and disease.


Subject(s)
Immunity, Innate , Intestinal Diseases/immunology , Intestines/immunology , Lymphoid Tissue/immunology , Metagenome , Animals , Humans , Intestinal Diseases/microbiology , Intestines/microbiology , Lymphoid Tissue/microbiology
20.
Immunity ; 37(1): 158-70, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22705104

ABSTRACT

Signals from commensal bacteria can influence immune cell development and susceptibility to infectious or inflammatory diseases. However, the mechanisms by which commensal bacteria regulate protective immunity after exposure to systemic pathogens remain poorly understood. Here, we demonstrate that antibiotic-treated (ABX) mice exhibit impaired innate and adaptive antiviral immune responses and substantially delayed viral clearance after exposure to systemic LCMV or mucosal influenza virus. Furthermore, ABX mice exhibited severe bronchiole epithelial degeneration and increased host mortality after influenza virus infection. Genome-wide transcriptional profiling of macrophages isolated from ABX mice revealed decreased expression of genes associated with antiviral immunity. Moreover, macrophages from ABX mice exhibited defective responses to type I and type II IFNs and impaired capacity to limit viral replication. Collectively, these data indicate that commensal-derived signals provide tonic immune stimulation that establishes the activation threshold of the innate immune system required for optimal antiviral immunity.


Subject(s)
Bacteria/immunology , Immunity, Innate , Viruses/immunology , Adaptive Immunity , Animals , Anti-Bacterial Agents/pharmacology , Arenaviridae Infections/genetics , Arenaviridae Infections/immunology , Bacteria/drug effects , Disease Susceptibility/immunology , Interferons/immunology , Lymphocytic choriomeningitis virus/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology
SELECTION OF CITATIONS
SEARCH DETAIL