Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Cell ; 176(3): 610-624.e18, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30612739

ABSTRACT

Plasma cells (PC) are found in the CNS of multiple sclerosis (MS) patients, yet their source and role in MS remains unclear. We find that some PC in the CNS of mice with experimental autoimmune encephalomyelitis (EAE) originate in the gut and produce immunoglobulin A (IgA). Moreover, we show that IgA+ PC are dramatically reduced in the gut during EAE, and likewise, a reduction in IgA-bound fecal bacteria is seen in MS patients during disease relapse. Removal of plasmablast (PB) plus PC resulted in exacerbated EAE that was normalized by the introduction of gut-derived IgA+ PC. Furthermore, mice with an over-abundance of IgA+ PB and/or PC were specifically resistant to the effector stage of EAE, and expression of interleukin (IL)-10 by PB plus PC was necessary and sufficient to confer resistance. Our data show that IgA+ PB and/or PC mobilized from the gut play an unexpected role in suppressing neuroinflammation.


Subject(s)
Immunoglobulin A/metabolism , Interleukin-10/metabolism , Intestines/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Immunoglobulin A/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Neuroimmunomodulation/immunology , Plasma Cells/metabolism
2.
Immunity ; 55(7): 1148-1150, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35830822

ABSTRACT

A plethora of studies have established the importance of eosinophils in protective immunity against infections and in allergy. In this issue of Immunity, Ignacio et al. (2022) define a vital for eosinophils in coordinating a microbiota-epithelial-immune axis that maintains intestinal homeostasis.


Subject(s)
Intestines , Microbiota , Eosinophils , Homeostasis
4.
Nat Immunol ; 17(6): 656-65, 2016 06.
Article in English | MEDLINE | ID: mdl-27043409

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) regulate tissue inflammation and repair after activation by cell-extrinsic factors such as host-derived cytokines. However, the cell-intrinsic metabolic pathways that control ILC2 function are undefined. Here we demonstrate that expression of the enzyme arginase-1 (Arg1) during acute or chronic lung inflammation is a conserved trait of mouse and human ILC2s. Deletion of mouse ILC-intrinsic Arg1 abrogated type 2 lung inflammation by restraining ILC2 proliferation and dampening cytokine production. Mechanistically, inhibition of Arg1 enzymatic activity disrupted multiple components of ILC2 metabolic programming by altering arginine catabolism, impairing polyamine biosynthesis and reducing aerobic glycolysis. These data identify Arg1 as a key regulator of ILC2 bioenergetics that controls proliferative capacity and proinflammatory functions promoting type 2 inflammation.


Subject(s)
Arginase/metabolism , Lymphocytes/physiology , Pneumonia/immunology , Animals , Arginase/genetics , Cell Proliferation/genetics , Cells, Cultured , Cytokines/metabolism , Glycolysis/genetics , Humans , Immunity, Innate , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Polyamines/metabolism , Th2 Cells/immunology
5.
Immunol Rev ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890777

ABSTRACT

The human gut microbiome is well-recognized as a key player in maintaining health. However, it is a dynamic entity that changes across the lifespan. How the microbial changes that occur in later decades of life shape host health or impact age-associated inflammatory neurological diseases such as multiple sclerosis (MS) is still unclear. Current understanding of the aging gut microbiome is largely limited to cross-sectional observational studies. Moreover, studies in humans are limited by confounding host-intrinsic and extrinsic factors that are not easily disentangled from aging. This review provides a comprehensive summary of existing literature on the aging gut microbiome and its known relationships with neurological diseases, with a specific focus on MS. We will also discuss preclinical animal models and human studies that shed light on the complex microbiota-host interactions that have the potential to influence disease pathology and progression in aging individuals. Lastly, we propose potential avenues of investigation to deconvolute features of an aging microbiota that contribute to disease, or alternatively promote health in advanced age.

6.
Immunity ; 49(4): 709-724.e8, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30291028

ABSTRACT

B cells thwart antigenic aggressions by releasing immunoglobulin M (IgM), IgG, IgA, and IgE, which deploy well-understood effector functions. In contrast, the role of secreted IgD remains mysterious. We found that some B cells generated IgD-secreting plasma cells following early exposure to external soluble antigens such as food proteins. Secreted IgD targeted basophils by interacting with the CD44-binding protein galectin-9. When engaged by antigen, basophil-bound IgD increased basophil secretion of interleukin-4 (IL-4), IL-5, and IL-13, which facilitated the generation of T follicular helper type 2 cells expressing IL-4. These germinal center T cells enhanced IgG1 and IgE but not IgG2a and IgG2b responses to the antigen initially recognized by basophil-bound IgD. In addition, IgD ligation by antigen attenuated allergic basophil degranulation induced by IgE co-ligation. Thus, IgD may link B cells with basophils to optimize humoral T helper type 2-mediated immunity against common environmental soluble antigens.


Subject(s)
Basophils/immunology , Galectins/immunology , Hyaluronan Receptors/immunology , Immunoglobulin D/immunology , Th2 Cells/immunology , Animals , Basophils/metabolism , Cell Line, Tumor , Cells, Cultured , Galectins/genetics , Galectins/metabolism , Gene Expression Profiling/methods , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Immunoglobulin D/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Mice, Inbred BALB C , Protein Binding , Th2 Cells/metabolism
7.
Nat Immunol ; 15(10): 929-937, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25151491

ABSTRACT

The paradigm that macrophages that reside in steady-state tissues are derived from embryonic precursors has never been investigated in the intestine, which contains the largest pool of macrophages. Using fate-mapping models and monocytopenic mice, together with bone marrow chimera and parabiotic models, we found that embryonic precursor cells seeded the intestinal mucosa and demonstrated extensive in situ proliferation during the neonatal period. However, these cells did not persist in the intestine of adult mice. Instead, they were replaced around the time of weaning by the chemokine receptor CCR2-dependent influx of Ly6C(hi) monocytes that differentiated locally into mature, anti-inflammatory macrophages. This process was driven largely by the microbiota and had to be continued throughout adult life to maintain a normal intestinal macrophage pool.


Subject(s)
Intestinal Mucosa/immunology , Intestines/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Animals, Newborn , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , Antigens, Ly/immunology , Antigens, Ly/metabolism , Bone Marrow Transplantation , CD11b Antigen/genetics , CD11b Antigen/immunology , CD11b Antigen/metabolism , CX3C Chemokine Receptor 1 , Cell Differentiation/immunology , Cell Proliferation , Flow Cytometry , Gene Expression/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestines/cytology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological , Monocytes/metabolism , Parabiosis , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Receptors, CCR2/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
8.
Immunity ; 42(2): 216-226, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25692699

ABSTRACT

Type 2 inflammatory responses can be elicited by diverse stimuli, including toxins, venoms, allergens, and infectious agents, and play critical roles in resistance and tolerance associated with infection, wound healing, tissue repair, and tumor development. Emerging data suggest that in addition to characteristic type 2-associated cytokines, the epidermal growth factor (EGF)-like molecule Amphiregulin (AREG) might be a critical component of type 2-mediated resistance and tolerance. Notably, numerous studies demonstrate that in addition to the established role of epithelial- and mesenchymal-derived AREG, multiple leukocyte populations including mast cells, basophils, group 2 innate lymphoid cells (ILC2s), and a subset of tissue-resident regulatory CD4(+) T cells can express AREG. In this review, we discuss recent advances in our understanding of the AREG-EGF receptor pathway and its involvement in infection and inflammation and propose a model for the function of this pathway in the context of resistance and tissue tolerance.


Subject(s)
EGF Family of Proteins/immunology , ErbB Receptors/immunology , Immune Tolerance/immunology , Inflammation/immunology , Wound Healing/immunology , Amphiregulin , Animals , Helminthiasis/immunology , Humans , Influenza, Human/immunology , Mice , Neoplasms/immunology , Orthomyxoviridae Infections/immunology , Regeneration , Tumor Escape/immunology
9.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34625492

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) control the formation of intestinal lymphoid tissues and play key roles in intestinal defense. They express neuropeptide vasoactive intestinal peptide (VIP) receptor 2 (VPAC2), through which VIP modulates their function, but whether VIP exerts other effects on ILC3 remains unclear. We show that VIP promotes ILC3 recruitment to the intestine through VPAC1 independent of the microbiota or adaptive immunity. VIP is also required for postnatal formation of lymphoid tissues as well as the maintenance of local populations of retinoic acid (RA)-producing dendritic cells, with RA up-regulating gut-homing receptor CCR9 expression by ILC3s. Correspondingly, mice deficient in VIP or VPAC1 suffer a paucity of intestinal ILC3s along with impaired production of the cytokine IL-22, rendering them highly susceptible to the enteric pathogen Citrobacter rodentium This heightened susceptibility to C. rodentium infection was ameliorated by RA supplementation, adoptive transfer of ILC3s, or by recombinant IL-22. Thus, VIP regulates the recruitment of intestinal ILC3s and formation of postnatal intestinal lymphoid tissues, offering protection against enteric pathogens.


Subject(s)
Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Lymphocytes/immunology , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Dendritic Cells/immunology , Gastrointestinal Microbiome/immunology , Interleukins/analysis , Lymphoid Tissue/cytology , Lymphoid Tissue/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR/biosynthesis , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Tretinoin/metabolism , Vasoactive Intestinal Peptide/genetics , Interleukin-22
10.
J Virol ; 96(6): e0206521, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35107369

ABSTRACT

Recent evidence indicates that viral components of the microbiota can contribute to intestinal homeostasis and protection from local inflammatory or infectious insults. However, host-derived mechanisms that regulate the virome remain largely unknown. In this study, we used colonization with the model commensal murine norovirus (MNV; strain CR6) to interrogate host-directed mechanisms of viral regulation, and we show that STAT1 is a central coordinator of both viral replication and antiviral T cell responses. In addition to restricting CR6 replication to the intestinal tract, we show that STAT1 regulates antiviral CD4+ and CD8+ T cell responses and prevents systemic viral-induced tissue damage and disease. Despite altered T cell responses that resemble those that mediate lethal immunopathology in systemic viral infections in STAT1-deficient mice, depletion of adaptive immune cells and their associated effector functions had no effect on CR6-induced disease. However, therapeutic administration of an antiviral compound limited viral replication, preventing virus-induced tissue damage and death without impacting the generation of inflammatory antiviral T cell responses. Collectively, our data show that STAT1 restricts MNV CR6 replication within the intestinal mucosa and that uncontrolled viral replication mediates disease rather than the concomitant development of dysregulated antiviral T cell responses in STAT1-deficient mice. IMPORTANCE The intestinal microbiota is a collection of bacteria, archaea, fungi, and viruses that colonize the mammalian gut. Coevolution of the host and microbiota has required development of immunological tolerance to prevent ongoing inflammatory responses against intestinal microbes. Breakdown of tolerance to bacterial components of the microbiota can contribute to immune activation and inflammatory disease. However, the mechanisms that are necessary to maintain tolerance to viral components of the microbiome, and the consequences of loss of tolerance, are less well understood. Here, we show that STAT1 is integral for preventing escape of a commensal-like virus, murine norovirus CR6 (MNV CR6), from the gut and that in the absence of STAT1, mice succumb to infection-induced disease. In contrast to the case with other systemic viral infections, mortality of STAT1-deficient mice is not driven by immune-mediated pathology. Our data demonstrate the importance of host-mediated geographical restriction of commensal-like viruses.


Subject(s)
Caliciviridae Infections , Norovirus , STAT1 Transcription Factor , T-Lymphocytes , Virus Replication , Animals , Caliciviridae Infections/mortality , Caliciviridae Infections/physiopathology , Intestinal Mucosa/virology , Mice , Norovirus/physiology , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , T-Lymphocytes/immunology , T-Lymphocytes/virology
11.
Immunity ; 39(6): 1158-70, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24332033

ABSTRACT

Extramedullary hematopoiesis (EMH) refers to the differentiation of hematopoietic stem cells (HSCs) into effector cells that occurs in compartments outside of the bone marrow. Previous studies linked pattern-recognition receptor (PRR)-expressing HSCs, EMH, and immune responses to microbial stimuli. However, whether EMH operates in broader immune contexts remains unknown. Here, we demonstrate a previously unrecognized role for thymic stromal lymphopoietin (TSLP) in promoting the population expansion of progenitor cells in the periphery and identify that TSLP-elicited progenitors differentiated into effector cells including macrophages, dendritic cells, and granulocytes and that these cells contributed to type 2 cytokine responses. The frequency of circulating progenitor cells was also increased in allergic patients with a gain-of-function polymorphism in TSLP, suggesting the TSLP-EMH pathway might operate in human disease. These data identify that TSLP-induced EMH contributes to the development of allergic inflammation and indicate that EMH is a conserved mechanism of innate immunity.


Subject(s)
Cytokines/metabolism , Hematopoiesis, Extramedullary/immunology , Hypersensitivity/immunology , Inflammation , Spleen/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Flow Cytometry , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Polymorphism, Genetic , Precursor Cells, B-Lymphoid/cytology , Spleen/cytology , Trichinellosis/immunology , Thymic Stromal Lymphopoietin
12.
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
13.
Mult Scler ; 26(12): 1448-1458, 2020 10.
Article in English | MEDLINE | ID: mdl-31971074

ABSTRACT

Helminthic worms are ancestral members of the intestinal ecosystem that have been largely eradicated from the general population in industrialized countries. Immunomodulatory mechanisms induced by some helminths mediate a "truce" between the mammalian host and the colonizing worm, thus allowing for long-term persistence in the absence of immune-mediated collateral tissue damage. This concept and the geographic discrepancy between global burdens of chronic inflammatory diseases and helminth infection have sparked interest in the potential of using helminthic worms as a therapeutic intervention to limit the progression of autoimmune diseases such as multiple sclerosis (MS). Here, we present and evaluate the evidence for this hypothesis in the pre-clinical animal model of MS, experimental autoimmune encephalitis, in helminth-infected MS patients and in clinical trials of administered helminth immunotherapy (HIT).


Subject(s)
Helminthiasis , Helminths , Multiple Sclerosis , Animals , Ecosystem , Helminthiasis/therapy , Humans , Immunotherapy , Multiple Sclerosis/therapy
15.
Nature ; 504(7478): 153-7, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24185009

ABSTRACT

The development and severity of inflammatory bowel diseases and other chronic inflammatory conditions can be influenced by host genetic and environmental factors, including signals derived from commensal bacteria. However, the mechanisms that integrate these diverse cues remain undefined. Here we demonstrate that mice with an intestinal epithelial cell (IEC)-specific deletion of the epigenome-modifying enzyme histone deacetylase 3 (HDAC3(ΔIEC) mice) exhibited extensive dysregulation of IEC-intrinsic gene expression, including decreased basal expression of genes associated with antimicrobial defence. Critically, conventionally housed HDAC3(ΔIEC) mice demonstrated loss of Paneth cells, impaired IEC function and alterations in the composition of intestinal commensal bacteria. In addition, HDAC3(ΔIEC) mice showed significantly increased susceptibility to intestinal damage and inflammation, indicating that epithelial expression of HDAC3 has a central role in maintaining intestinal homeostasis. Re-derivation of HDAC3(ΔIEC) mice into germ-free conditions revealed that dysregulated IEC gene expression, Paneth cell homeostasis and intestinal barrier function were largely restored in the absence of commensal bacteria. Although the specific mechanisms through which IEC-intrinsic HDAC3 expression regulates these complex phenotypes remain to be determined, these data indicate that HDAC3 is a critical factor that integrates commensal-bacteria-derived signals to calibrate epithelial cell responses required to establish normal host-commensal relationships and maintain intestinal homeostasis.


Subject(s)
Gene Expression Regulation , Histone Deacetylases/metabolism , Homeostasis , Intestinal Mucosa/enzymology , Intestines/microbiology , Symbiosis , Adult , Animals , Bacteria/genetics , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Crohn Disease/enzymology , Crohn Disease/genetics , Crohn Disease/microbiology , Female , Gene Deletion , Gene Expression Profiling , Histone Deacetylases/genetics , Humans , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Paneth Cells/cytology , Paneth Cells/metabolism , RNA, Ribosomal, 16S/genetics , Signal Transduction
16.
PLoS Pathog ; 12(6): e1005684, 2016 06.
Article in English | MEDLINE | ID: mdl-27327515

ABSTRACT

In order for a virus to persist, there must be a balance between viral replication and immune clearance. It is commonly believed that adaptive immunity drives clearance of viral infections and, thus, dysfunction or viral evasion of adaptive immunity is required for a virus to persist. Type I interferons (IFNs) play pleiotropic roles in the antiviral response, including through innate control of viral replication. Murine norovirus (MNoV) replicates in dendritic cells (DCs) and type I IFN signaling in DCs is important for early control of MNoV replication. We show here that the non-persistent MNoV strain CW3 persists systemically when CD11c positive DCs are unable to respond to type I IFN. Persistence in this setting is associated with increased early viral titers, maintenance of DC numbers, increased expression of DC activation markers and an increase in CD8 T cell and antibody responses. Furthermore, CD8 T cell function is maintained during the persistent phase of infection and adaptive immune cells from persistently infected mice are functional when transferred to Rag1-/- recipients. Finally, increased early replication and persistence are also observed in mixed bone marrow chimeras where only half of the CD11c positive DCs are unable to respond to type I IFN. These findings demonstrate that increased early viral replication due to a cell-intrinsic innate immune deficiency is sufficient for persistence and a functional adaptive immune response is not sufficient for viral clearance.


Subject(s)
Caliciviridae Infections/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Interferon Type I/immunology , Receptor, Interferon alpha-beta/immunology , Adaptive Immunity/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Norovirus , Real-Time Polymerase Chain Reaction , Receptor, Interferon alpha-beta/deficiency , Virus Replication/physiology
17.
Proc Natl Acad Sci U S A ; 112(34): 10762-7, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26243875

ABSTRACT

The barrier surfaces of the skin, lung, and intestine are constantly exposed to environmental stimuli that can result in inflammation and tissue damage. Interleukin (IL)-33-dependent group 2 innate lymphoid cells (ILC2s) are enriched at barrier surfaces and have been implicated in promoting inflammation; however, the mechanisms underlying the tissue-protective roles of IL-33 or ILC2s at surfaces such as the intestine remain poorly defined. Here we demonstrate that, following activation with IL-33, expression of the growth factor amphiregulin (AREG) is a dominant functional signature of gut-associated ILC2s. In the context of a murine model of intestinal damage and inflammation, the frequency and number of AREG-expressing ILC2s increases following intestinal injury and genetic disruption of the endogenous AREG-epidermal growth factor receptor (EGFR) pathway exacerbated disease. Administration of exogenous AREG limited intestinal inflammation and decreased disease severity in both lymphocyte-sufficient and lymphocyte-deficient mice, revealing a previously unrecognized innate immune mechanism of intestinal tissue protection. Furthermore, treatment with IL-33 or transfer of ILC2s ameliorated intestinal disease severity in an AREG-dependent manner. Collectively, these data reveal a critical feedback loop in which cytokine cues from damaged epithelia activate innate immune cells to express growth factors essential for ILC-dependent restoration of epithelial barrier function and maintenance of tissue homeostasis.


Subject(s)
Colitis/immunology , EGF Family of Proteins/physiology , ErbB Receptors/physiology , Immunity, Innate/physiology , Immunity, Mucosal/physiology , Interleukin-33/physiology , Lymphocytes/immunology , Amphiregulin , Animals , Colitis/chemically induced , Colitis/therapy , Dextran Sulfate/toxicity , Disease Models, Animal , EGF Family of Proteins/deficiency , EGF Family of Proteins/therapeutic use , Epithelium/immunology , Epithelium/metabolism , Epithelium/pathology , Feedback, Physiological , Immunotherapy, Adoptive , Interleukin-33/biosynthesis , Interleukin-33/genetics , Interleukin-33/therapeutic use , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lung/immunology , Lung/pathology , Lymphocytes/classification , Mice , Mice, Knockout , Mucins/biosynthesis , Peyer's Patches/immunology , Peyer's Patches/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Proteins/therapeutic use , Signal Transduction , Specific Pathogen-Free Organisms
18.
J Immunol ; 190(5): 2292-300, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23355735

ABSTRACT

Resistin-like molecule (RELM)α belongs to a family of secreted mammalian proteins that have putative immunomodulatory functions. Recent studies have identified a pathogenic role for RELMα in chemically induced colitis through effects on innate cell populations. However, whether RELMα regulates intestinal adaptive immunity to enteric pathogens is unknown. In this study, we employed Citrobacter rodentium as a physiologic model of pathogenic Escherichia coli-induced diarrheal disease, colitis, and Th17 cell responses. In response to Citrobacter, RELMα expression was induced in intestinal epithelial cells, infiltrating macrophages, and eosinophils of the infected colons. Citrobacter-infected RELMα(-/-) mice exhibited reduced infection-induced intestinal inflammation, characterized by decreased leukocyte recruitment to the colons and reduced immune cell activation compared with wild-type (WT) mice. Interestingly, Citrobacter colonization and clearance were unaffected in RELMα(-/-) mice, suggesting that the immune stimulatory effects of RELMα following Citrobacter infection were pathologic rather than host-protective. Furthermore, infected RELMα(-/-) mice exhibited decreased CD4(+) T cell expression of the proinflammatory cytokine IL-17A. To directly test whether RELMα promoted Citrobacter-induced intestinal inflammation via IL-17A, infected WT and IL-17A(-/-) mice were treated with rRELMα. RELMα treatment of Citrobacter-infected WT mice exacerbated intestinal inflammation and IL-17A expression whereas IL-17A(-/-) mice were protected from RELMα-induced intestinal inflammation. Finally, infected RELMα(-/-) mice exhibited reduced levels of serum IL-23p19 compared with WT mice, and RELMα(-/-) peritoneal macrophages showed deficient IL-23p19 induction. Taken together, these data identify a proinflammatory role for RELMα in bacterial-induced colitis and suggest that the IL-23/Th17 axis is a critical mediator of RELMα-induced inflammation.


Subject(s)
Citrobacter rodentium/immunology , Inflammation/immunology , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-17/immunology , Intestines/drug effects , Macrophages, Peritoneal/drug effects , Th17 Cells/drug effects , Adaptive Immunity/drug effects , Animals , Citrobacter rodentium/pathogenicity , Dextran Sulfate , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/pathology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Gene Expression/drug effects , Inflammation/chemically induced , Inflammation/microbiology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-23 Subunit p19/blood , Interleukin-23 Subunit p19/immunology , Intestines/immunology , Intestines/microbiology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Th17 Cells/immunology , Th17 Cells/pathology
19.
J Virol ; 87(12): 7015-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23596300

ABSTRACT

Norovirus (NV) gastroenteritis is a major contributor to global morbidity and mortality, yet little is known about immune mechanisms leading to NV control. Previous studies using the murine norovirus (MNV) model have established a key role for T cells in MNV clearance. Despite these advances, important questions remain regarding the magnitude, location, and dynamics of the MNV-specific T cell response. To address these questions, we identified MNV-specific major histocompatibility complex (MHC) class I immunodominant epitopes using an overlapping peptide screen. One of these epitopes (amino acids 519 to 527 of open reading frame 2 [ORF2(519-527)]) was highly conserved among all NV genogroups. Using MHC class I peptide tetramers, we tracked MNV-specific CD8 T cells in lymphoid and mucosal sites during infection with two MNV strains with distinct biological behaviors, the acutely cleared strain CW3 and the persistent strain CR6. Here, we show that enteric MNV infection elicited robust T cell responses primarily in the intestinal mucosa and that MNV-specific CD8 T cells dynamically regulated the expression of surface molecules associated with activation, differentiation, and homing. Furthermore, compared to MNV-CW3 infection, chronic infection with MNV-CR6 resulted in fewer and less-functional CD8 T cells, and this difference was evident as early as day 8 postinfection. Finally, MNV-specific CD8 T cells were capable of reducing the viral load in persistently infected Rag1(-/-) mice, suggesting that these cells are a crucial component of NV immunity. Collectively, these data provide fundamental new insights into the adaptive immune response to two closely related NV strains with distinct biological behaviors and bring us closer to understanding the correlates of protective antiviral immunity in the intestine.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Caliciviridae Infections/immunology , Gastroenteritis/immunology , Intestinal Mucosa , Norovirus/immunology , Animals , Caliciviridae Infections/virology , Chronic Disease , Epitopes, T-Lymphocyte , Female , Flow Cytometry , Gastroenteritis/virology , Immunodominant Epitopes , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Lymphocyte Activation , Major Histocompatibility Complex , Mice , Mice, Inbred C57BL
20.
J Vis Exp ; (203)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38345223

ABSTRACT

Bacteriophages (phages) are viruses that infect bacteria with species- and strain-level specificity and are the most abundant biological entities across all known ecosystems. Within bacterial communities, such as those found in the gut microbiota, phages are implicated in regulating microbiota population dynamics and driving bacterial evolution. There has been renewed interest in phage research in the last decade, in part due to the host-specific killing capabilities of lytic phages, which offer a promising tool to counter the increasing threat of antimicrobial resistant bacteria. Furthermore, recent studies demonstrating that phages adhere to intestinal mucus suggest they may have a protective role in preventing bacterial invasion into the underlying epithelium. Importantly, like bacterial microbiomes, disrupted phageomes have been associated with worsened outcomes in diseases such as inflammatory bowel disease. Previous studies have demonstrated that phages can modulate the microbiome of animals and humans through fecal filtrate transplants, benefiting the host's health. With this recent wave of research comes the necessity to establish and standardize protocols for studying phages in the context of the gut microbiome. This protocol provides a set of procedures to study isolated T4 phages and their bacterial host, Escherichia coli, in the context of the murine gastrointestinal tract. The methods described here outline how to start from a phage lysate, administer it to mice and assess effects on bacterial host and phage levels. This protocol can be modified and applied to other phage-bacterial pairs and provides a starting point for studying host-phage dynamics in vivo.


Subject(s)
Bacteriophages , Microbiota , Humans , Mice , Animals , Bacteriophages/physiology , Bacteriophage T4 , Escherichia coli , Gastrointestinal Tract/microbiology , Intestines , Bacteria
SELECTION OF CITATIONS
SEARCH DETAIL