Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Cell ; 162(5): 1078-89, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26317471

ABSTRACT

Regulatory T (Treg) cells suppress immune responses to a broad range of non-microbial and microbial antigens and indirectly limit immune inflammation-inflicted tissue damage by employing multiple mechanisms of suppression. Here, we demonstrate that selective Treg cell deficiency in amphiregulin leads to severe acute lung damage and decreased blood oxygen concentration during influenza virus infection without any measureable alterations in Treg cell suppressor function, antiviral immune responses, or viral load. This tissue repair modality is mobilized in Treg cells in response to inflammatory mediator IL-18 or alarmin IL-33, but not by TCR signaling that is required for suppressor function. These results suggest that, during infectious lung injury, Treg cells have a major direct and non-redundant role in tissue repair and maintenance-distinct from their role in suppression of immune responses and inflammation-and that these two essential Treg cell functions are invoked by separable cues.


Subject(s)
Influenza, Human/immunology , Lung/cytology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Amphiregulin/genetics , Animals , Autoimmunity , Disease Models, Animal , Humans , Influenza, Human/pathology , Lung/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Suppressor Factors, Immunologic/analysis , T-Lymphocytes, Regulatory/chemistry
2.
Cell ; 154(1): 213-27, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23827684

ABSTRACT

Bioactive lipid mediators play a crucial role in the induction and resolution of inflammation. To elucidate their involvement during influenza infection, liquid chromatography/mass spectrometry lipidomic profiling of 141 lipid species was performed on a mouse influenza model using two viruses of significantly different pathogenicity. Infection by the low-pathogenicity strain X31/H3N2 induced a proinflammatory response followed by a distinct anti-inflammatory response; infection by the high-pathogenicity strain PR8/H1N1 resulted in overlapping pro- and anti-inflammatory states. Integration of the large-scale lipid measurements with targeted gene expression data demonstrated that 5-lipoxygenase metabolites correlated with the pathogenic phase of the infection, whereas 12/15-lipoxygenase metabolites were associated with the resolution phase. Hydroxylated linoleic acid, specifically the ratio of 13- to 9-hydroxyoctadecadienoic acid, was identified as a potential biomarker for immune status during an active infection. Importantly, some of the findings from the animal model were recapitulated in studies of human nasopharyngeal lavages obtained during the 2009-2011 influenza seasons.


Subject(s)
Eicosanoids/isolation & purification , Fatty Acids, Unsaturated/isolation & purification , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/immunology , Lipids/analysis , Orthomyxoviridae Infections/immunology , Animals , Arachidonate 5-Lipoxygenase/metabolism , Cytokines/immunology , Disease Models, Animal , Eicosanoids/immunology , Fatty Acids, Unsaturated/immunology , Humans , Inflammation Mediators/analysis , Metabolic Networks and Pathways , Mice , Nasal Lavage Fluid/immunology , Transcriptome
3.
Cell ; 150(1): 29-38, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770213

ABSTRACT

Regulatory T (Treg) cells, whose differentiation and function are controlled by X chromosome-encoded transcription factor Foxp3, are generated in the thymus (tTreg) and extrathymically (peripheral, pTreg), and their deficiency results in fatal autoimmunity. Here, we demonstrate that a Foxp3 enhancer, conserved noncoding sequence 1 (CNS1), essential for pTreg but dispensable for tTreg cell generation, is present only in placental mammals. CNS1 is largely composed of mammalian-wide interspersed repeats (MIR) that have undergone retrotransposition during early mammalian radiation. During pregnancy, pTreg cells specific to a model paternal alloantigen were generated in a CNS1-dependent manner and accumulated in the placenta. Furthermore, when mated with allogeneic, but not syngeneic, males, CNS1-deficient females showed increased fetal resorption accompanied by increased immune cell infiltration and defective remodeling of spiral arteries. Our results suggest that, during evolution, a CNS1-dependent mechanism of extrathymic differentiation of Treg cells emerged in placental animals to enforce maternal-fetal tolerance.


Subject(s)
Immune Tolerance , Mammals/immunology , Placenta/cytology , Placenta/immunology , Pregnancy/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Enhancer Elements, Genetic , Female , Fetus/immunology , Forkhead Transcription Factors/genetics , Humans , Male , Mammals/genetics , Mice , Opossums
4.
Immunity ; 43(5): 933-44, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26588779

ABSTRACT

Mutations in ADAR, which encodes the ADAR1 RNA-editing enzyme, cause Aicardi-Goutières syndrome (AGS), a severe autoimmune disease associated with an aberrant type I interferon response. How ADAR1 prevents autoimmunity remains incompletely defined. Here, we demonstrate that ADAR1 is a specific and essential negative regulator of the MDA5-MAVS RNA sensing pathway. Moreover, we uncovered a MDA5-MAVS-independent function for ADAR1 in the development of multiple organs. We showed that the p150 isoform of ADAR1 uniquely regulated the MDA5 pathway, whereas both the p150 and p110 isoforms contributed to development. Abrupt deletion of ADAR1 in adult mice revealed that both of these functions were required throughout life. Our findings delineate genetically separable roles for both ADAR1 isoforms in vivo, with implications for the human diseases caused by ADAR mutations.


Subject(s)
Adenosine Deaminase/metabolism , Autoimmunity/physiology , DEAD-box RNA Helicases/metabolism , Protein Isoforms/metabolism , RNA Editing/physiology , RNA/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autoimmune Diseases of the Nervous System/metabolism , HEK293 Cells , Humans , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1 , Mice , Nervous System Malformations/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/physiology
6.
Nature ; 546(7658): 421-425, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28607488

ABSTRACT

Adaptive immune responses are tailored to different types of pathogens through differentiation of naive CD4 T cells into functionally distinct subsets of effector T cells (T helper 1 (TH1), TH2, and TH17) defined by expression of the key transcription factors T-bet, GATA3, and RORγt, respectively. Regulatory T (Treg) cells comprise a distinct anti-inflammatory lineage specified by the X-linked transcription factor Foxp3 (refs 2, 3). Paradoxically, some activated Treg cells express the aforementioned effector CD4 T cell transcription factors, which have been suggested to provide Treg cells with enhanced suppressive capacity. Whether expression of these factors in Treg cells-as in effector T cells-is indicative of heterogeneity of functionally discrete and stable differentiation states, or conversely may be readily reversible, is unknown. Here we demonstrate that expression of the TH1-associated transcription factor T-bet in mouse Treg cells, induced at steady state and following infection, gradually becomes highly stable even under non-permissive conditions. Loss of function or elimination of T-bet-expressing Treg cells-but not of T-bet expression in Treg cells-resulted in severe TH1 autoimmunity. Conversely, following depletion of T-bet- Treg cells, the remaining T-bet+ cells specifically inhibited TH1 and CD8 T cell activation consistent with their co-localization with T-bet+ effector T cells. These results suggest that T-bet+ Treg cells have an essential immunosuppressive function and indicate that Treg cell functional heterogeneity is a critical feature of immunological tolerance.


Subject(s)
Immune Tolerance/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Animals , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Separation , Female , Lymphocyte Activation , Male , Mice , T-Lymphocytes, Regulatory/cytology , Th1 Cells/cytology , Th17 Cells/cytology , Th17 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology
7.
Proc Natl Acad Sci U S A ; 117(27): 15789-15798, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32581129

ABSTRACT

Patients infected with influenza are at high risk of secondary bacterial infection, which is a major proximate cause of morbidity and mortality. We have shown that in mice, prior infection with influenza results in increased inflammation and mortality upon Staphylococcus aureus infection, recapitulating the human disease. Lipidomic profiling of the lungs of superinfected mice revealed an increase in CYP450 metabolites during lethal superinfection. These lipids are endogenous ligands for the nuclear receptor PPARα, and we demonstrate that Ppara-/- mice are less susceptible to superinfection than wild-type mice. PPARα is an inhibitor of NFκB activation, and transcriptional profiling of cells isolated by bronchoalveolar lavage confirmed that influenza infection inhibits NFκB, thereby dampening proinflammatory and prosurvival signals. Furthermore, network analysis indicated an increase in necrotic cell death in the lungs of superinfected mice compared to mice infected with S. aureus alone. Consistent with this, we observed reduced NFκB-mediated inflammation and cell survival signaling in cells isolated from the lungs of superinfected mice. The kinase RIPK3 is required to induce necrotic cell death and is strongly induced in cells isolated from the lungs of superinfected mice compared to mice infected with S. aureus alone. Genetic and pharmacological perturbations demonstrated that PPARα mediates RIPK3-dependent necroptosis and that this pathway plays a central role in mortality following superinfection. Thus, we have identified a molecular circuit in which infection with influenza induces CYP450 metabolites that activate PPARα, leading to increased necrotic cell death in the lung which correlates with the excess mortality observed in superinfection.


Subject(s)
Inflammation/genetics , Influenza, Human/genetics , PPAR alpha/genetics , Staphylococcal Infections/genetics , Superinfection/genetics , Animals , Bronchoalveolar Lavage/methods , Coinfection/genetics , Coinfection/microbiology , Coinfection/mortality , Cytochrome P-450 Enzyme System/genetics , Disease Models, Animal , Disease Susceptibility , Humans , Inflammation/microbiology , Inflammation/mortality , Influenza, Human/microbiology , Influenza, Human/mortality , Lung/microbiology , Lung/pathology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Knockout , Necroptosis/genetics , Staphylococcal Infections/microbiology , Staphylococcal Infections/mortality , Superinfection/mortality
8.
PLoS Pathog ; 16(1): e1008251, 2020 01.
Article in English | MEDLINE | ID: mdl-31961914

ABSTRACT

Patients with cystic fibrosis (CF) have altered fecal microbiomes compared to those of healthy controls. The magnitude of this dysbiosis correlates with measures of CF gastrointestinal (GI) disease, including GI inflammation and nutrient malabsorption. However, whether this dysbiosis is caused by mutations in the CFTR gene, the underlying defect in CF, or whether CF-associated dysbiosis augments GI disease was not clear. To test the relationships between CFTR dysfunction, microbes, and intestinal health, we established a germ-free (GF) CF mouse model and demonstrated that CFTR gene mutations are sufficient to alter the GI microbiome. Furthermore, flow cytometric analysis demonstrated that colonized CF mice have increased mesenteric lymph node and spleen TH17+ cells compared with non-CF mice, suggesting that CFTR defects alter adaptive immune responses. Our findings demonstrate that CFTR mutations modulate both the host adaptive immune response and the intestinal microbiome.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/microbiology , Dysbiosis/microbiology , Gastrointestinal Microbiome , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Cystic Fibrosis/genetics , Cystic Fibrosis/immunology , Cystic Fibrosis Transmembrane Conductance Regulator/immunology , Disease Models, Animal , Dysbiosis/genetics , Dysbiosis/immunology , Female , Humans , Intestines/immunology , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Mutation
9.
Nat Immunol ; 11(12): 1136-42, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21057511

ABSTRACT

Macrophages mediate crucial innate immune responses via caspase-1-dependent processing and secretion of interleukin 1ß (IL-1ß) and IL-18. Although infection with wild-type Salmonella typhimurium is lethal to mice, we show here that a strain that persistently expresses flagellin was cleared by the cytosolic flagellin-detection pathway through the activation of caspase-1 by the NLRC4 inflammasome; however, this clearance was independent of IL-1ß and IL-18. Instead, caspase-1-induced pyroptotic cell death released bacteria from macrophages and exposed the bacteria to uptake and killing by reactive oxygen species in neutrophils. Similarly, activation of caspase-1 cleared unmanipulated Legionella pneumophila and Burkholderia thailandensis by cytokine-independent mechanisms. This demonstrates that activation of caspase-1 clears intracellular bacteria in vivo independently of IL-1ß and IL-18 and establishes pyroptosis as an efficient mechanism of bacterial clearance by the innate immune system.


Subject(s)
Apoptosis/immunology , Caspase 1/immunology , Immunity, Innate/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Animals , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Inflammasomes/immunology , Inflammasomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL
10.
Int J Obes (Lond) ; 45(7): 1542-1552, 2021 07.
Article in English | MEDLINE | ID: mdl-33934107

ABSTRACT

BACKGROUND: Retinoic acid (RA) controls diverse physiological functions including weight regulation and energy metabolism. It has been reported that mice lacking ALDH1A1, one of the aldehyde dehydrogenases (ALDH) that synthesize RA, are healthy and resistant to weight gain, raising the possibility that inhibiting this enzyme might treat obesity. We previously demonstrated that treatment with a pan-ALDH1A enzyme inhibitor, WIN18446, suppressed weight gain in mice fed a high-fat diet (HFD), but caused increased hepatic lipidosis and reversible male infertility. METHODS: A series of piperazine compounds that inhibited ALDH1A1 were identified and their inhibitory activity was characterized in vitro using purified recombinant enzymes and cell-based assay systems. One potent compound, FSI-TN42 (N42) was examined for its oral bioavailability and pharmacodynamic effects. In addition, its effect on weight gain was investigated by daily oral administration to C57BL/6 male mice receiving a HFD, and compared with mice receiving WIN18446 or vehicle alone (n = 6/group, 200 mg compound/kg body weight) for 5 weeks. Body weights were measured weekly, and a glucose tolerance test was performed after 4 weeks of treatment. Tissues were collected to determine changes in adipose weight, hepatic lipidosis, retinoid metabolism, and expression of genes associated with RA and lipid metabolism. RESULTS: N42 irreversibly binds and inhibits ALDH1A1 in vitro with a low nM IC50 and 800-fold specificity for ALDH1A1 compared to ALDH1A2. Daily oral administration of N42 significantly suppressed weight gain (P < 0.05) and reduced visceral adiposity (p < 0.05) in mice fed a HFD without the hepatic lipidosis observed with WIN18446 treatment. CONCLUSIONS: We developed a potent and specific inhibitor of ALDH1A1 that suppressed weight gain in mice fed a HFD. These findings demonstrate that inhibition of ALDH1A1 is a feasible target for drug development to treat and/or prevent obesity.


Subject(s)
Aldehyde Dehydrogenase 1 Family/antagonists & inhibitors , Obesity/metabolism , Piperazines/pharmacology , Retinal Dehydrogenase/antagonists & inhibitors , Weight Gain/drug effects , Adipose Tissue/drug effects , Administration, Oral , Animals , Diet, High-Fat , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Piperazines/administration & dosage , Piperazines/chemistry
11.
PLoS Pathog ; 15(8): e1007899, 2019 08.
Article in English | MEDLINE | ID: mdl-31415679

ABSTRACT

West Nile Virus (WNV), an emerging and re-emerging RNA virus, is the leading source of arboviral encephalitic morbidity and mortality in the United States. WNV infections are acutely controlled by innate immunity in peripheral tissues outside of the central nervous system (CNS) but WNV can evade the actions of interferon (IFN) to facilitate CNS invasion, causing encephalitis, encephalomyelitis, and death. Recent studies indicate that STimulator of INterferon Gene (STING), canonically known for initiating a type I IFN production and innate immune response to cytosolic DNA, is required for host defense against neurotropic RNA viruses. We evaluated the role of STING in host defense to control WNV infection and pathology in a murine model of infection. When challenged with WNV, STING knock out (-/-) mice displayed increased morbidity and mortality compared to wild type (WT) mice. Virologic analysis and assessment of STING activation revealed that STING signaling was not required for control of WNV in the spleen nor was WNV sufficient to mediate canonical STING activation in vitro. However, STING-/- mice exhibited a clear trend of increased viral load and virus dissemination in the CNS. We found that STING-/- mice exhibited increased and prolonged neurological signs compared to WT mice. Pathological examination revealed increased lesions, mononuclear cellular infiltration and neuronal death in the CNS of STING-/- mice, with sustained pathology after viral clearance. We found that STING was required in bone marrow derived macrophages for early control of WNV replication and innate immune activation. In vivo, STING-/- mice developed an aberrant T cell response in both the spleen and brain during WNV infection that linked with increased and sustained CNS pathology compared to WT mice. Our findings demonstrate that STING plays a critical role in immune programming for the control of neurotropic WNV infection and CNS disease.


Subject(s)
Central Nervous System/immunology , Central Nervous System/pathology , Immunity, Innate/immunology , Membrane Proteins/physiology , Virus Replication , West Nile Fever/immunology , West Nile virus/immunology , Animals , Central Nervous System/metabolism , Central Nervous System/virology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Viral Load , West Nile Fever/metabolism , West Nile Fever/virology
12.
Nat Immunol ; 10(11): 1170-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19767756

ABSTRACT

The transcription factor Foxp3 has an indispensable role in establishing stable transcriptional and functional programs of regulatory T cells (T(reg) cells). Loss of Foxp3 expression in mature T(reg) cells results in a failure of suppressor function, yet the molecular mechanisms that ensure steady, heritable Foxp3 expression in the T(reg) cell lineage remain unknown. Using T(reg) cell-specific gene targeting, we found that complexes of the transcription factors Runx and CBFbeta were required for maintenance of Foxp3 mRNA and protein expression in T(reg) cells. Consequently, mice lacking CBFbetab exclusively in the T(reg) cell lineage had a moderate lymphoproliferative syndrome. Thus, Runx-CBFbeta complexes maintain stable high expression of Foxp3 and serve as an essential determinant of T(reg) cell lineage stability.


Subject(s)
Core Binding Factor beta Subunit/immunology , Forkhead Transcription Factors/immunology , Gene Expression Regulation , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Bone Marrow Transplantation , Cell Lineage/immunology , Core Binding Factor beta Subunit/metabolism , Female , Forkhead Transcription Factors/metabolism , Gene Targeting , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Male , Mice , Mice, Inbred C57BL , Spleen/cytology , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology , Thymus Gland/immunology
13.
Immunity ; 36(1): 120-31, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22284419

ABSTRACT

The type I interferon (IFN) response initiated by detection of nucleic acids is important for antiviral defense but is also associated with specific autoimmune diseases. Mutations in the human 3' repair exonuclease 1 (Trex1) gene cause Aicardi-Goutières syndrome (AGS), an IFN-associated autoimmune disease. However, the source of the type I IFN response and the precise mechanisms of disease in AGS remain unknown. Here, we demonstrate that Trex1 is an essential negative regulator of the STING-dependent antiviral response. We used an in vivo reporter of IFN activity in Trex1-deficient mice to localize the initiation of disease to nonhematopoietic cells. These IFNs drove T cell-mediated inflammation and an autoantibody response that targeted abundant, tissue-restricted autoantigens. However, B cells contributed to mortality independently of T cell-mediated tissue damage. These findings reveal a stepwise progression of autoimmune disease in Trex1-deficient mice, with implications for the treatment of AGS and related disorders.


Subject(s)
Autoimmune Diseases/physiopathology , Autoimmunity/immunology , Interferons , Lymphocytes/immunology , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases of the Nervous System/physiopathology , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Interferons/physiology , Mice , Mice, Knockout , Models, Biological , Nervous System Malformations/physiopathology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Signal Transduction
14.
Nature ; 528(7580): 132-136, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26605529

ABSTRACT

T-cell receptor (TCR) signalling has a key role in determining T-cell fate. Precursor cells expressing TCRs within a certain low-affinity range for complexes of self-peptide and major histocompatibility complex (MHC) undergo positive selection and differentiate into naive T cells expressing a highly diverse self-MHC-restricted TCR repertoire. In contrast, precursors displaying TCRs with a high affinity for 'self' are either eliminated through TCR-agonist-induced apoptosis (negative selection) or restrained by regulatory T (Treg) cells, whose differentiation and function are controlled by the X-chromosome-encoded transcription factor Foxp3 (reviewed in ref. 2). Foxp3 is expressed in a fraction of self-reactive T cells that escape negative selection in response to agonist-driven TCR signals combined with interleukin 2 (IL-2) receptor signalling. In addition to Treg cells, TCR-agonist-driven selection results in the generation of several other specialized T-cell lineages such as natural killer T cells and innate mucosal-associated invariant T cells. Although the latter exhibit a restricted TCR repertoire, Treg cells display a highly diverse collection of TCRs. Here we explore in mice whether a specialized mechanism enables agonist-driven selection of Treg cells with a diverse TCR repertoire, and the importance this holds for self-tolerance. We show that the intronic Foxp3 enhancer conserved noncoding sequence 3 (CNS3) acts as an epigenetic switch that confers a poised state to the Foxp3 promoter in precursor cells to make Treg cell lineage commitment responsive to a broad range of TCR stimuli, particularly to suboptimal ones. CNS3-dependent expansion of the TCR repertoire enables Treg cells to control self-reactive T cells effectively, especially when thymic negative selection is genetically impaired. Our findings highlight the complementary roles of these two main mechanisms of self-tolerance.


Subject(s)
Self Tolerance/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Cell Lineage , Conserved Sequence/genetics , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Female , Forkhead Transcription Factors/genetics , Introns/genetics , Male , Mice , Promoter Regions, Genetic/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin-2/immunology , Receptors, Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/deficiency , AIRE Protein
15.
Immunity ; 34(4): 566-78, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21511185

ABSTRACT

Effector CD4+ T cell subsets, whose differentiation is facilitated by distinct cytokine cues, amplify the corresponding type of inflammatory response. Regulatory T (Treg) cells integrate environmental cues to suppress particular types of inflammation. In this regard, STAT3, a transcription factor essential for T helper 17 (Th17) cell differentiation, is necessary for Treg cell-mediated control of Th17 cell responses. Here, we showed that anti-inflammatory interleukin-10 (IL-10), and not proinflammatory IL-6 and IL-23 cytokine signaling, endowed Treg cells with the ability to suppress pathogenic Th17 cell responses. Ablation of the IL-10 receptor in Treg cells resulted in selective dysregulation of Th17 cell responses and colitis similar to that observed in mice harboring STAT3-deficient Treg cells. Thus, Treg cells limit Th17 cell inflammation by serving as principal amplifiers of negative regulatory circuits operating in immune effector cells.


Subject(s)
Colitis/immunology , Interleukin-10/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cell Lineage , Colitis/pathology , Interleukin-10/metabolism , Mice , Mice, Knockout , Phosphorylation , Receptors, Interleukin-10/deficiency , Receptors, Interleukin-10/immunology , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/cytology
16.
Nature ; 515(7527): 365-70, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25409825

ABSTRACT

The basic body plan and major physiological axes have been highly conserved during mammalian evolution, yet only a small fraction of the human genome sequence appears to be subject to evolutionary constraint. To quantify cis- versus trans-acting contributions to mammalian regulatory evolution, we performed genomic DNase I footprinting of the mouse genome across 25 cell and tissue types, collectively defining ∼8.6 million transcription factor (TF) occupancy sites at nucleotide resolution. Here we show that mouse TF footprints conjointly encode a regulatory lexicon that is ∼95% similar with that derived from human TF footprints. However, only ∼20% of mouse TF footprints have human orthologues. Despite substantial turnover of the cis-regulatory landscape, nearly half of all pairwise regulatory interactions connecting mouse TF genes have been maintained in orthologous human cell types through evolutionary innovation of TF recognition sequences. Furthermore, the higher-level organization of mouse TF-to-TF connections into cellular network architectures is nearly identical with human. Our results indicate that evolutionary selection on mammalian gene regulation is targeted chiefly at the level of trans-regulatory circuitry, enabling and potentiating cis-regulatory plasticity.


Subject(s)
Conserved Sequence/genetics , Evolution, Molecular , Mammals/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , DNA Footprinting , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Humans , Mice
17.
J Pharmacol Exp Ther ; 371(1): 25-35, 2019 10.
Article in English | MEDLINE | ID: mdl-31375638

ABSTRACT

Methylcellulose (MC; 0.5% concentration) is commonly used when evaluating investigational agents for efficacy in preclinical models of disease. When administered by the oral (PO) route, MC is considered a Food and Drug Administration "generally recognized as safe" compound. Yet, there is limited data pertaining to the tolerability and impact on model fidelity of repeated intraperitoneal administration of 0.5% MC. Chronic administration of high-concentration MC (2%-2.5%) has been used to induce anemia, splenomegaly, and lesions in multiple organ systems in several preclinical species. Histopathological findings from a diagnostic pathologic analysis of a single mouse from our laboratory with experimentally induced chronic seizures that had received repeated intraperitoneal administration of antiseizure drugs delivered in MC revealed similar widespread lesions. This study thus tested the hypothesis that chronic administration of intraperitoneal, but not PO, MC incites histologic lesions without effects on preclinical phenotype. Male CF-1 mice (n = 2-14/group) were randomized to receive either 6 weeks of twice weekly 0.5% MC or saline (intraperitoneal or PO) following induction of chronic seizures. Histology of a subset of mice revealed lesions in kidney, liver, mediastinal lymph nodes, mesentery, aorta, and choroid plexus only in intraperitoneal MC-treated mice (n = 7/7). Kindled mice that received MC PO (n = 5) or saline (intraperitoneal n = 6, PO n = 3) had no lesions. There were no effects of intraperitoneal MC treatment on body weight, appearance, seizure stability, or behavior. Nonetheless, our findings suggest that repeated intraperitoneal, but not PO, MC elicits systemic organ damage without impacting the model phenotype, which may confound interpretation of investigational drug-induced histologic lesions. SIGNIFICANCE STATEMENT: Methylcellulose (0.5% concentration) is commonly used when evaluating investigational agents for efficacy in preclinical models of disease. Herein, we demonstrate that repeated administration of 0.5% methylcellulose by the intraperitoneal, but not oral, route results in systemic inflammation and presence of foam-laden macrophages but does not impact the behavioral phenotype of a rodent model of neurological disease.


Subject(s)
Injections, Intraperitoneal/adverse effects , Methylcellulose/adverse effects , Phenotype , Seizures/chemically induced , Animals , Aorta/drug effects , Choroid Plexus/drug effects , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Female , Kidney/drug effects , Liver/drug effects , Lymph Nodes/drug effects , Male , Methylcellulose/administration & dosage , Methylcellulose/toxicity , Mice , Mice, Inbred C57BL
18.
PLoS Pathog ; 13(4): e1006305, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28380049

ABSTRACT

Antiviral responses must rapidly defend against infection while minimizing inflammatory damage, but the mechanisms that regulate the magnitude of response within an infected cell are not well understood. miRNAs are small non-coding RNAs that suppress protein levels by binding target sequences on their cognate mRNA. Here, we identify miR-144 as a negative regulator of the host antiviral response. Ectopic expression of miR-144 resulted in increased replication of three RNA viruses in primary mouse lung epithelial cells: influenza virus, EMCV, and VSV. We identified the transcriptional network regulated by miR-144 and demonstrate that miR-144 post-transcriptionally suppresses TRAF6 levels. In vivo ablation of miR-144 reduced influenza virus replication in the lung and disease severity. These data suggest that miR-144 reduces the antiviral response by attenuating the TRAF6-IRF7 pathway to alter the cellular antiviral transcriptional landscape.


Subject(s)
Influenza, Human/immunology , MicroRNAs/metabolism , Orthomyxoviridae/genetics , Signal Transduction , TNF Receptor-Associated Factor 6/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Animals , Cell Line , Epithelial Cells/virology , Gene Expression Profiling , Genes, Reporter , Humans , Influenza, Human/virology , Lung/virology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Orthomyxoviridae/immunology , Orthomyxoviridae/physiology , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , TNF Receptor-Associated Factor 6/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Viral Load , Virus Replication
19.
Am J Pathol ; 188(7): 1536-1554, 2018 07.
Article in English | MEDLINE | ID: mdl-29753791

ABSTRACT

Murine norovirus (MNV) is an RNA virus that can prove lethal in mice with impaired innate immunity. We found that MNV-4 infection of Stat1-/- mice was not lethal, but produced a 100% penetrant, previously undescribed lymphatic phenotype characterized by chronic-active lymphangitis with hepatitis, splenitis, and chronic cecal and colonic inflammation. Lesion pathogenesis progressed from early ileal enteritis and regional dilated lymphatics to lymphangitis, granulomatous changes in the liver and spleen, and, ultimately, typhlocolitis. Lesion development was neither affected by antibiotics nor reproduced by infection with another enteric RNA virus, rotavirus. MNV-4 infection in Stat1-/- mice decreased expression of vascular endothelial growth factor (Vegf) receptor 3, Vegf-c, and Vegf-d and increased interferon (Ifn)-γ, tumor necrosis factor-α, and inducible nitric oxide synthase. However, anti-IFN-γ and anti-tumor necrosis factor-α antibody treatment did not attenuate the histologic lesions. Studies in Ifnαßγr-/- mice suggested that canonical signaling via interferon receptors did not cause MNV-4-induced disease. Infected Stat1-/- mice had increased STAT3 phosphorylation and expressed many STAT3-regulated genes, consistent with our findings of increased myeloid cell subsets and serum granulocyte colony-stimulating factor, which are also associated with increased STAT3 activity. In conclusion, in Stat1-/- mice, MNV-4 induces lymphatic lesions similar to those seen in Crohn disease as well as hepatitis, splenitis, and typhlocolitis. MNV-4-infected Stat1-/- mice may be a useful model to study mechanistic associations between viral infections, lymphatic dysfunction, and intestinal inflammation in a genetically susceptible host.


Subject(s)
Caliciviridae Infections/complications , Colitis/pathology , Intestines/pathology , Liver/pathology , Lymphangitis/pathology , STAT1 Transcription Factor/physiology , Spleen/pathology , Animals , Caliciviridae Infections/virology , Colitis/metabolism , Colitis/virology , Female , Interferons/metabolism , Intestines/virology , Liver/metabolism , Liver/virology , Lymphangitis/metabolism , Lymphangitis/virology , Mice , Mice, Knockout , Norovirus/isolation & purification , Signal Transduction , Spleen/metabolism , Spleen/virology
20.
PLoS Pathog ; 12(11): e1005996, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27806117

ABSTRACT

Infection with West Nile virus (WNV) leads to a range of disease outcomes, including chronic infection, though lack of a robust mouse model of chronic WNV infection has precluded identification of the immune events contributing to persistent infection. Using the Collaborative Cross, a population of recombinant inbred mouse strains with high levels of standing genetic variation, we have identified a mouse model of persistent WNV disease, with persistence of viral loads within the brain. Compared to lines exhibiting no disease or marked disease, the F1 cross CC(032x013)F1 displays a strong immunoregulatory signature upon infection that correlates with restraint of the WNV-directed cytolytic response. We hypothesize that this regulatory T cell response sufficiently restrains the immune response such that a chronic infection can be maintained in the CNS. Use of this new mouse model of chronic neuroinvasive virus will be critical in developing improved strategies to prevent prolonged disease in humans.


Subject(s)
T-Lymphocytes, Regulatory/immunology , West Nile Fever/immunology , Animals , Chronic Disease , Disease Models, Animal , Female , Flow Cytometry , Humans , Male , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , West Nile virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL