Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Cell ; 185(19): 3501-3519.e20, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36041436

RESUMO

How intestinal microbes regulate metabolic syndrome is incompletely understood. We show that intestinal microbiota protects against development of obesity, metabolic syndrome, and pre-diabetic phenotypes by inducing commensal-specific Th17 cells. High-fat, high-sugar diet promoted metabolic disease by depleting Th17-inducing microbes, and recovery of commensal Th17 cells restored protection. Microbiota-induced Th17 cells afforded protection by regulating lipid absorption across intestinal epithelium in an IL-17-dependent manner. Diet-induced loss of protective Th17 cells was mediated by the presence of sugar. Eliminating sugar from high-fat diets protected mice from obesity and metabolic syndrome in a manner dependent on commensal-specific Th17 cells. Sugar and ILC3 promoted outgrowth of Faecalibaculum rodentium that displaced Th17-inducing microbiota. These results define dietary and microbiota factors posing risk for metabolic syndrome. They also define a microbiota-dependent mechanism for immuno-pathogenicity of dietary sugar and highlight an elaborate interaction between diet, microbiota, and intestinal immunity in regulation of metabolic disorders.


Assuntos
Síndrome Metabólica , Microbiota , Animais , Dieta Hiperlipídica , Açúcares da Dieta , Interleucina-17 , Mucosa Intestinal , Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Obesidade , Células Th17
2.
Cell ; 185(16): 2879-2898.e24, 2022 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-35931020

RESUMO

Human gut commensals are increasingly suggested to impact non-communicable diseases, such as inflammatory bowel diseases (IBD), yet their targeted suppression remains a daunting unmet challenge. In four geographically distinct IBD cohorts (n = 537), we identify a clade of Klebsiella pneumoniae (Kp) strains, featuring a unique antibiotics resistance and mobilome signature, to be strongly associated with disease exacerbation and severity. Transfer of clinical IBD-associated Kp strains into colitis-prone, germ-free, and colonized mice enhances intestinal inflammation. Stepwise generation of a lytic five-phage combination, targeting sensitive and resistant IBD-associated Kp clade members through distinct mechanisms, enables effective Kp suppression in colitis-prone mice, driving an attenuated inflammation and disease severity. Proof-of-concept assessment of Kp-targeting phages in an artificial human gut and in healthy volunteers demonstrates gastric acid-dependent phage resilience, safety, and viability in the lower gut. Collectively, we demonstrate the feasibility of orally administered combination phage therapy in avoiding resistance, while effectively inhibiting non-communicable disease-contributing pathobionts.


Assuntos
Bacteriófagos , Colite , Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais , Animais , Colite/terapia , Humanos , Inflamação/terapia , Doenças Inflamatórias Intestinais/terapia , Klebsiella pneumoniae , Camundongos
3.
Cell ; 163(2): 367-80, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26411289

RESUMO

Intestinal Th17 cells are induced and accumulate in response to colonization with a subgroup of intestinal microbes such as segmented filamentous bacteria (SFB) and certain extracellular pathogens. Here, we show that adhesion of microbes to intestinal epithelial cells (ECs) is a critical cue for Th17 induction. Upon monocolonization of germ-free mice or rats with SFB indigenous to mice (M-SFB) or rats (R-SFB), M-SFB and R-SFB showed host-specific adhesion to small intestinal ECs, accompanied by host-specific induction of Th17 cells. Citrobacter rodentium and Escherichia coli O157 triggered similar Th17 responses, whereas adhesion-defective mutants of these microbes failed to do so. Moreover, a mixture of 20 bacterial strains, which were selected and isolated from fecal samples of a patient with ulcerative colitis on the basis of their ability to cause a robust induction of Th17 cells in the mouse colon, also exhibited EC-adhesive characteristics.


Assuntos
Aderência Bacteriana , Citrobacter rodentium/fisiologia , Infecções por Enterobacteriaceae/imunologia , Infecções por Escherichia coli/imunologia , Escherichia coli O157/fisiologia , Mucosa Intestinal/imunologia , Células Th17/imunologia , Animais , Infecções Bacterianas/imunologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Células Epiteliais/ultraestrutura , Fezes/microbiologia , Humanos , Imunoglobulina A/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Microscopia Eletrônica de Varredura , Ratos , Ratos Endogâmicos F344 , Especificidade da Espécie
4.
Nature ; 629(8013): 901-909, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38658756

RESUMO

The liver is the main gateway from the gut, and the unidirectional sinusoidal flow from portal to central veins constitutes heterogenous zones, including the periportal vein (PV) and the pericentral vein zones1-5. However, functional differences in the immune system in each zone remain poorly understood. Here intravital imaging revealed that inflammatory responses are suppressed in PV zones. Zone-specific single-cell transcriptomics detected a subset of immunosuppressive macrophages enriched in PV zones that express high levels of interleukin-10 and Marco, a scavenger receptor that sequesters pro-inflammatory pathogen-associated molecular patterns and damage-associated molecular patterns, and consequently suppress immune responses. Induction of Marco+ immunosuppressive macrophages depended on gut microbiota. In particular, a specific bacterial family, Odoribacteraceae, was identified to induce this macrophage subset through its postbiotic isoallolithocholic acid. Intestinal barrier leakage resulted in inflammation in PV zones, which was markedly augmented in Marco-deficient conditions. Chronic liver inflammatory diseases such as primary sclerosing cholangitis (PSC) and non-alcoholic steatohepatitis (NASH) showed decreased numbers of Marco+ macrophages. Functional ablation of Marco+ macrophages led to PSC-like inflammatory phenotypes related to colitis and exacerbated steatosis in NASH in animal experimental models. Collectively, commensal bacteria induce Marco+ immunosuppressive macrophages, which consequently limit excessive inflammation at the gateway of the liver. Failure of this self-limiting system promotes hepatic inflammatory disorders such as PSC and NASH.


Assuntos
Colangite Esclerosante , Microbioma Gastrointestinal , Inflamação , Fígado , Macrófagos , Hepatopatia Gordurosa não Alcoólica , Simbiose , Animais , Feminino , Humanos , Masculino , Camundongos , Bacteroidetes/metabolismo , Colangite Esclerosante/imunologia , Colangite Esclerosante/microbiologia , Colangite Esclerosante/patologia , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Perfilação da Expressão Gênica , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/patologia , Interleucina-10/imunologia , Interleucina-10/metabolismo , Fígado/imunologia , Fígado/patologia , Fígado/microbiologia , Macrófagos/citologia , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/imunologia , Hepatopatia Gordurosa não Alcoólica/microbiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Veia Porta , Receptores Imunológicos/deficiência , Receptores Imunológicos/metabolismo , Análise de Célula Única , Simbiose/imunologia
5.
Nature ; 609(7927): 582-589, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36071157

RESUMO

Increased levels of proteases, such as trypsin, in the distal intestine have been implicated in intestinal pathological conditions1-3. However, the players and mechanisms that underlie protease regulation in the intestinal lumen have remained unclear. Here we show that Paraprevotella strains isolated from the faecal microbiome of healthy human donors are potent trypsin-degrading commensals. Mechanistically, Paraprevotella recruit trypsin to the bacterial surface through type IX secretion system-dependent polysaccharide-anchoring proteins to promote trypsin autolysis. Paraprevotella colonization protects IgA from trypsin degradation and enhances the effectiveness of oral vaccines against Citrobacter rodentium. Moreover, Paraprevotella colonization inhibits lethal infection with murine hepatitis virus-2, a mouse coronavirus that is dependent on trypsin and trypsin-like proteases for entry into host cells4,5. Consistently, carriage of putative genes involved in trypsin degradation in the gut microbiome was associated with reduced severity of diarrhoea in patients with SARS-CoV-2 infection. Thus, trypsin-degrading commensal colonization may contribute to the maintenance of intestinal homeostasis and protection from pathogen infection.


Assuntos
Microbioma Gastrointestinal , Intestino Grosso , Simbiose , Tripsina , Administração Oral , Animais , Sistemas de Secreção Bacterianos , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Bacteroidetes/isolamento & purificação , Bacteroidetes/metabolismo , COVID-19/complicações , Citrobacter rodentium/imunologia , Diarreia/complicações , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Humanos , Imunoglobulina A/metabolismo , Intestino Grosso/metabolismo , Intestino Grosso/microbiologia , Camundongos , Vírus da Hepatite Murina/metabolismo , Vírus da Hepatite Murina/patogenicidade , Proteólise , SARS-CoV-2/patogenicidade , Tripsina/metabolismo , Internalização do Vírus
6.
Nature ; 599(7885): 458-464, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34325466

RESUMO

Centenarians have a decreased susceptibility to ageing-associated illnesses, chronic inflammation and infectious diseases1-3. Here we show that centenarians have a distinct gut microbiome that is enriched in microorganisms that are capable of generating unique secondary bile acids, including various isoforms of lithocholic acid (LCA): iso-, 3-oxo-, allo-, 3-oxoallo- and isoallolithocholic acid. Among these bile acids, the biosynthetic pathway for isoalloLCA had not been described previously. By screening 68 bacterial isolates from the faecal microbiota of a centenarian, we identified Odoribacteraceae strains as effective producers of isoalloLCA both in vitro and in vivo. Furthermore, we found that the enzymes 5α-reductase (5AR) and 3ß-hydroxysteroid dehydrogenase (3ß-HSDH) were responsible for the production of isoalloLCA. IsoalloLCA exerted potent antimicrobial effects against Gram-positive (but not Gram-negative) multidrug-resistant pathogens, including Clostridioides difficile and Enterococcus faecium. These findings suggest that the metabolism of specific bile acids may be involved in reducing the risk of infection with pathobionts, thereby potentially contributing to the maintenance of intestinal homeostasis.


Assuntos
Bactérias/metabolismo , Vias Biossintéticas , Centenários , Microbioma Gastrointestinal , Ácido Litocólico/análogos & derivados , Ácido Litocólico/biossíntese , 3-Hidroxiesteroide Desidrogenases/metabolismo , Idoso de 80 Anos ou mais , Animais , Antibacterianos/biossíntese , Antibacterianos/metabolismo , Bactérias/classificação , Bactérias/enzimologia , Bactérias/isolamento & purificação , Colestenona 5 alfa-Redutase/metabolismo , Fezes/química , Fezes/microbiologia , Feminino , Bactérias Gram-Positivas/metabolismo , Humanos , Ácido Litocólico/metabolismo , Masculino , Camundongos , Simbiose
7.
Nat Immunol ; 15(6): 571-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24777532

RESUMO

Intestinal regulatory T cells (Treg cells) are necessary for the suppression of excessive immune responses to commensal bacteria. However, the molecular machinery that controls the homeostasis of intestinal Treg cells has remained largely unknown. Here we report that colonization of germ-free mice with gut microbiota upregulated expression of the DNA-methylation adaptor Uhrf1 in Treg cells. Mice with T cell-specific deficiency in Uhrf1 (Uhrf1(fl/fl)Cd4-Cre mice) showed defective proliferation and functional maturation of colonic Treg cells. Uhrf1 deficiency resulted in derepression of the gene (Cdkn1a) that encodes the cyclin-dependent kinase inhibitor p21 due to hypomethylation of its promoter region, which resulted in cell-cycle arrest of Treg cells. As a consequence, Uhrf1(fl/fl)Cd4-Cre mice spontaneously developed severe colitis. Thus, Uhrf1-dependent epigenetic silencing of Cdkn1a was required for the maintenance of gut immunological homeostasis. This mechanism enforces symbiotic host-microbe interactions without an inflammatory response.


Assuntos
Colite/imunologia , Colo/imunologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Epigênese Genética , Proteínas Nucleares/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Proteínas Estimuladoras de Ligação a CCAAT , Pontos de Checagem do Ciclo Celular , Proliferação de Células , Células Cultivadas , Clostridium/imunologia , Colite/genética , Colo/microbiologia , Metilação de DNA , Perfilação da Expressão Gênica , Interleucina-2 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microbiota/imunologia , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Simbiose/imunologia , Ubiquitina-Proteína Ligases , Regulação para Cima
8.
Nat Immunol ; 14(3): 281-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23334788

RESUMO

TCRαß thymocytes differentiate into either CD8αß(+) cytotoxic T lymphocytes or CD4(+) helper T cells. This functional dichotomy is controlled by key transcription factors, including the helper T cell master regulator ThPOK, which suppresses the cytolytic program in major histocompatibility complex (MHC) class II-restricted CD4(+) thymocytes. ThPOK continues to repress genes of the CD8 lineage in mature CD4(+) T cells, even as they differentiate into effector helper T cell subsets. Here we found that the helper T cell fate was not fixed and that mature, antigen-stimulated CD4(+) T cells terminated expression of the gene encoding ThPOK and reactivated genes of the CD8 lineage. This unexpected plasticity resulted in the post-thymic termination of the helper T cell program and the functional differentiation of distinct MHC class II-restricted CD4(+) cytotoxic T lymphocytes.


Assuntos
Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Citrobacter rodentium/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Proteínas de Homeodomínio/genética , Interleucina-7/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/metabolismo , Timócitos/metabolismo
9.
Nature ; 565(7741): 600-605, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30675064

RESUMO

There is a growing appreciation for the importance of the gut microbiota as a therapeutic target in various diseases. However, there are only a handful of known commensal strains that can potentially be used to manipulate host physiological functions. Here we isolate a consortium of 11 bacterial strains from healthy human donor faeces that is capable of robustly inducing interferon-γ-producing CD8 T cells in the intestine. These 11 strains act together to mediate the induction without causing inflammation in a manner that is dependent on CD103+ dendritic cells and major histocompatibility (MHC) class Ia molecules. Colonization of mice with the 11-strain mixture enhances both host resistance against Listeria monocytogenes infection and the therapeutic efficacy of immune checkpoint inhibitors in syngeneic tumour models. The 11 strains primarily represent rare, low-abundance components of the human microbiome, and thus have great potential as broadly effective biotherapeutics.


Assuntos
Adenocarcinoma/imunologia , Adenocarcinoma/terapia , Bactérias/classificação , Linfócitos T CD8-Positivos/imunologia , Microbioma Gastrointestinal/imunologia , Listeriose/prevenção & controle , Simbiose/imunologia , Adenocarcinoma/patologia , Animais , Antígenos CD/metabolismo , Bactérias/imunologia , Bactérias/isolamento & purificação , Linfócitos T CD8-Positivos/citologia , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Fezes/microbiologia , Feminino , Voluntários Saudáveis , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Cadeias alfa de Integrinas/metabolismo , Interferon gama/biossíntese , Interferon gama/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/microbiologia , Masculino , Camundongos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Nat Immunol ; 13(7): 659-66, 2012 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-22610141

RESUMO

Although the mechanisms by which innate pathogen-recognition receptors enhance adaptive immune responses are increasingly well understood, whether signaling events from distinct classes of receptors affect each other in modulating adaptive immunity remains unclear. We found here that the activation of cytosolic RIG-I-like receptors (RLRs) resulted in the selective suppression of transcription of the gene encoding the p40 subunit of interleukin 12 (Il12b) that was effectively induced by the activation of Toll-like receptors (TLRs). The RLR-activated transcription factor IRF3 bound dominantly, relative to IRF5, to the Il12b promoter, where it interfered with the TLR-induced assembly of a productive transcription-factor complex. The activation of RLRs in mice attenuated TLR-induced responses of the T helper type 1 cell (T(H)1 cell) and interleukin 17-producing helper T cell (T(H)17 cell) subset types and, consequently, viral infection of mice caused death at sublethal doses of bacterial infection. The innate immune receptor cross-interference we describe may have implications for infection-associated clinical episodes.


Assuntos
Transdução de Sinais/imunologia , Linfócitos T/imunologia , Receptores Toll-Like/imunologia , Sequência de Aminoácidos , Animais , Infecções Bacterianas/imunologia , Células Cultivadas , Regulação da Expressão Gênica/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fatores Reguladores de Interferon/metabolismo , Subunidade p40 da Interleucina-12/metabolismo , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Células Th1/imunologia , Células Th17/imunologia , Fatores de Transcrição/metabolismo , Viroses/imunologia
11.
Mol Psychiatry ; 28(5): 1932-1945, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36882500

RESUMO

The BTBR T+Itpr3tf/J (BTBR/J) strain is one of the most valid models of idiopathic autism, serving as a potent forward genetics tool to dissect the complexity of autism. We found that a sister strain with an intact corpus callosum, BTBR TF/ArtRbrc (BTBR/R), showed more prominent autism core symptoms but moderate ultrasonic communication/normal hippocampus-dependent memory, which may mimic autism in the high functioning spectrum. Intriguingly, disturbed epigenetic silencing mechanism leads to hyperactive endogenous retrovirus (ERV), a mobile genetic element of ancient retroviral infection, which increases de novo copy number variation (CNV) formation in the two BTBR strains. This feature makes the BTBR strain a still evolving multiple-loci model toward higher ASD susceptibility. Furthermore, active ERV, analogous to virus infection, evades the integrated stress response (ISR) of host defense and hijacks the transcriptional machinery during embryonic development in the BTBR strains. These results suggest dual roles of ERV in the pathogenesis of ASD, driving host genome evolution at a long-term scale and managing cellular pathways in response to viral infection, which has immediate effects on embryonic development. The wild-type Draxin expression in BTBR/R also makes this substrain a more precise model to investigate the core etiology of autism without the interference of impaired forebrain bundles as in BTBR/J.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Retrovirus Endógenos , Gravidez , Feminino , Humanos , Animais , Camundongos , Retrovirus Endógenos/genética , Variações do Número de Cópias de DNA , Transtorno Autístico/etiologia , Prosencéfalo/metabolismo , Corpo Caloso/patologia , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/complicações , Camundongos Endogâmicos
12.
Cell ; 139(3): 485-98, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19836068

RESUMO

The gastrointestinal tract of mammals is inhabited by hundreds of distinct species of commensal microorganisms that exist in a mutualistic relationship with the host. How commensal microbiota influence the host immune system is poorly understood. We show here that colonization of the small intestine of mice with a single commensal microbe, segmented filamentous bacterium (SFB), is sufficient to induce the appearance of CD4(+) T helper cells that produce IL-17 and IL-22 (Th17 cells) in the lamina propria. SFB adhere tightly to the surface of epithelial cells in the terminal ileum of mice with Th17 cells but are absent from mice that have few Th17 cells. Colonization with SFB was correlated with increased expression of genes associated with inflammation and antimicrobial defenses and resulted in enhanced resistance to the intestinal pathogen Citrobacter rodentium. Thus, manipulation of this commensal-regulated pathway may provide new opportunities for enhancing mucosal immunity and treating autoimmune disease.


Assuntos
Bactérias Gram-Positivas/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Diferenciação Celular , Citrobacter rodentium/imunologia , Bactérias Gram-Positivas/fisiologia , Imunidade nas Mucosas/imunologia , Interleucina-17/imunologia , Interleucinas/imunologia , Mucosa Intestinal/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Mucosa/imunologia , Mucosa/microbiologia , Proteína Amiloide A Sérica/metabolismo , Organismos Livres de Patógenos Específicos , Simbiose , Interleucina 22
13.
Nat Immunol ; 12(5): 450-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21460847

RESUMO

The immunoregulatory cytokine interleukin 10 (IL-10) is expressed mainly by T helper type 2 (T(H)2) cells but also by T(H)1 cells during chronic infection. Here we observed plasticity in the expression of IL-10 and IL-13 after chronic T(H)1 stimulation; furthermore, the expression of Il10 and Il13 was regulated by the transcription factor E4BP4. Chronically stimulated E4BP4-deficient (Nfil3(-/-); called 'E4bp4(-/-)' here) T(H)1 cells, regulatory T cells (T(reg) cells) and natural killer T cells (NKT cells) had attenuated expression of IL-10 and IL-13. Enforced expression of E4bp4 initiated the production of IL-10 and IL-13 by conventional T(H)1 cells. E4bp4(-/-) T(H)2 cells showed impairment of IL-10 production with no effect on IL-13. Our results indicate that E4BP4 has multiple functions in controlling the plasticity of IL-13 in T(H)1 cells and IL-10 in T(H)1 cells, T(H)2 cells, T(reg) cells and NKT cells.


Assuntos
Doenças Autoimunes/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Linfócitos T CD4-Positivos/imunologia , Interleucina-10/imunologia , Interleucina-13/imunologia , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Regiões Promotoras Genéticas , Organismos Livres de Patógenos Específicos , Transcrição Gênica
14.
Mol Psychiatry ; 27(8): 3343-3354, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35491410

RESUMO

Immune dysregulation plays a key role in the pathogenesis of autism. Changes occurring at the systemic level, from brain inflammation to disturbed innate/adaptive immune in the periphery, are frequently observed in patients with autism; however, the intrinsic mechanisms behind them remain elusive. We hypothesize a common etiology may lie in progenitors of different types underlying widespread immune dysregulation. By single-cell RNA sequencing (sc-RNA seq), we trace the developmental origins of immune dysregulation in a mouse model of idiopathic autism. It is found that both in aorta-gonad-mesonephros (AGM) and yolk sac (YS) progenitors, the dysregulation of HDAC1-mediated epigenetic machinery alters definitive hematopoiesis during embryogenesis and downregulates the expression of the AP-1 complex for microglia development. Subsequently, these changes result in the dysregulation of the immune system, leading to gut dysbiosis and hyperactive microglia in the brain. We further confirm that dysregulated immune profiles are associated with specific microbiota composition, which may serve as a biomarker to identify autism of immune-dysregulated subtypes. Our findings elucidate a shared mechanism for the origin of immune dysregulation from the brain to the gut in autism and provide new insight to dissecting the heterogeneity of autism, as well as the therapeutic potential of targeting immune-dysregulated autism subtypes.


Assuntos
Transtorno Autístico , Camundongos , Animais , Transtorno Autístico/genética , Mesonefro , Saco Vitelino/fisiologia , Gônadas , Epigênese Genética/genética , Modelos Animais de Doenças
15.
Immunity ; 41(1): 152-65, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25017466

RESUMO

Foxp3(+) T cells play a critical role for the maintenance of immune tolerance. Here we show that in mice, Foxp3(+) T cells contributed to diversification of gut microbiota, particularly of species belonging to Firmicutes. The control of indigenous bacteria by Foxp3(+) T cells involved regulatory functions both outside and inside germinal centers (GCs), consisting of suppression of inflammation and regulation of immunoglobulin A (IgA) selection in Peyer's patches, respectively. Diversified and selected IgAs contributed to maintenance of diversified and balanced microbiota, which in turn facilitated the expansion of Foxp3(+) T cells, induction of GCs, and IgA responses in the gut through a symbiotic regulatory loop. Thus, the adaptive immune system, through cellular and molecular components that are required for immune tolerance and through the diversification as well as selection of antibody repertoire, mediates host-microbial symbiosis by controlling the richness and balance of bacterial communities required for homeostasis.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Imunoglobulina A/imunologia , Microbiota/imunologia , Imunidade Adaptativa , Animais , Fatores de Transcrição Forkhead/imunologia , Vida Livre de Germes , Centro Germinativo/imunologia , Proteínas de Homeodomínio/genética , Homeostase/imunologia , Tolerância Imunológica/imunologia , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID/microbiologia , Nódulos Linfáticos Agregados/imunologia , Simbiose/imunologia
16.
Nature ; 549(7673): 528-532, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28902840

RESUMO

Maternal immune activation (MIA) contributes to behavioural abnormalities associated with neurodevelopmental disorders in both primate and rodent offspring. In humans, epidemiological studies suggest that exposure of fetuses to maternal inflammation increases the likelihood of developing autism spectrum disorder. In pregnant mice, interleukin-17a (IL-17a) produced by T helper 17 (TH17) cells (CD4+ T helper effector cells involved in multiple inflammatory conditions) induces behavioural and cortical abnormalities in the offspring exposed to MIA. However, it is unclear whether other maternal factors are required to promote MIA-associated phenotypes. Moreover, the underlying mechanisms by which MIA leads to T cell activation with increased IL-17a in the maternal circulation are not well understood. Here we show that MIA phenotypes in offspring require maternal intestinal bacteria that promote TH17 cell differentiation. Pregnant mice that had been colonized with mouse commensal segmented filamentous bacteria or human commensal bacteria that induce intestinal TH17 cells were more likely to produce offspring with MIA-associated abnormalities. We also show that small intestine dendritic cells from pregnant, but not from non-pregnant, females secrete IL-1ß, IL-23 and IL-6 and stimulate T cells to produce IL-17a upon exposure to MIA. Overall, our data suggest that defined gut commensal bacteria with a propensity to induce TH17 cells may increase the risk of neurodevelopmental disorders in the offspring of pregnant mothers undergoing immune system activation owing to infections or autoinflammatory syndromes.


Assuntos
Microbioma Gastrointestinal/imunologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Animais , Comportamento Animal , Células Dendríticas/imunologia , Feminino , Inflamação/imunologia , Inflamação/microbiologia , Interleucina-17/imunologia , Interleucina-1beta/imunologia , Interleucina-23/imunologia , Interleucina-6/imunologia , Intestino Delgado/citologia , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Masculino , Camundongos , Fenótipo , Gravidez , Simbiose , Células Th17/citologia , Células Th17/imunologia
17.
Immunity ; 39(3): 584-98, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-24035363

RESUMO

Because immune responses simultaneously defend and injure the host, the immune system must be finely regulated to ensure the host's survival. Here, we have shown that when injected with high Toll-like receptor ligand doses or infected with lymphocytic choriomeningitis virus (LCMV) clone 13, which has a high viral turnover, inflammatory monocyte-derived dendritic cells (Mo-DCs) engulfed apoptotic erythroid cells. In this process, called hemophagocytosis, phosphatidylserine (PS) served as an "eat-me" signal. Type I interferons were necessary for both PS exposure on erythroid cells and the expression of PS receptors in the Mo-DCs. Importantly, hemophagocytosis was required for interleukin-10 (IL-10) production from Mo-DCs. Blocking hemophagocytosis or Mo-DC-derived IL-10 significantly increased cytotoxic T cell lymphocyte activity, tissue damage, and mortality in virus-infected hosts, suggesting that hemophagocytosis moderates immune responses to ensure the host's survival in vivo. This sheds light on the physiological relevance of hemophagocytosis in severe inflammatory and infectious diseases.


Assuntos
Apoptose/imunologia , Células Dendríticas/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Fagocitose , Animais , Diferenciação Celular , Células Dendríticas/metabolismo , Células Eritroides/imunologia , Interferon Tipo I/metabolismo , Interleucina-10/biossíntese , Ativação Linfocitária , Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Fosfatidilserinas/metabolismo , Receptores de Superfície Celular/metabolismo , Linfócitos T Citotóxicos/imunologia
18.
Immunity ; 38(5): 970-83, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23706669

RESUMO

Mouse and human dendritic cells (DCs) are composed of functionally specialized subsets, but precise interspecies correlation is currently incomplete. Here, we showed that murine lung and gut lamina propria CD11b+ DC populations were comprised of two subsets: FLT3- and IRF4-dependent CD24(+)CD64(-) DCs and contaminating CSF-1R-dependent CD24(-)CD64(+) macrophages. Functionally, loss of CD24(+)CD11b(+) DCs abrogated CD4+ T cell-mediated interleukin-17 (IL-17) production in steady state and after Aspergillus fumigatus challenge. Human CD1c+ DCs, the equivalent of murine CD24(+)CD11b(+) DCs, also expressed IRF4, secreted IL-23, and promoted T helper 17 cell responses. Our data revealed heterogeneity in the mouse CD11b+ DC compartment and identifed mucosal tissues IRF4-expressing DCs specialized in instructing IL-17 responses in both mouse and human. The demonstration of mouse and human DC subsets specialized in driving IL-17 responses highlights the conservation of key immune functions across species and will facilitate the translation of mouse in vivo findings to advance DC-based clinical therapies.


Assuntos
Aspergillus fumigatus/imunologia , Células Dendríticas/metabolismo , Fatores Reguladores de Interferon/metabolismo , Interleucina-17/metabolismo , Células Th17/metabolismo , Animais , Antígeno CD11b/metabolismo , Antígeno CD24/metabolismo , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Humanos , Interleucina-17/biossíntese , Interleucina-23/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Macrófagos/metabolismo , Camundongos , Receptores de IgG/metabolismo , Mucosa Respiratória/citologia , Mucosa Respiratória/imunologia , Tirosina Quinase 3 Semelhante a fms/metabolismo
19.
PLoS Pathog ; 14(4): e1006955, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29621339

RESUMO

Macrolides are used to treat various inflammatory diseases owing to their immunomodulatory properties; however, little is known about their precise mechanism of action. In this study, we investigated the functional significance of the expansion of myeloid-derived suppressor cell (MDSC)-like CD11b+Gr-1+ cells in response to the macrolide antibiotic clarithromycin (CAM) in mouse models of shock and post-influenza pneumococcal pneumonia as well as in humans. Intraperitoneal administration of CAM markedly expanded splenic and lung CD11b+Gr-1+ cell populations in naïve mice. Notably, CAM pretreatment enhanced survival in a mouse model of lipopolysaccharide (LPS)-induced shock. In addition, adoptive transfer of CAM-treated CD11b+Gr-1+ cells protected mice against LPS-induced lethality via increased IL-10 expression. CAM also improved survival in post-influenza, CAM-resistant pneumococcal pneumonia, with improved lung pathology as well as decreased interferon (IFN)-γ and increased IL-10 levels. Adoptive transfer of CAM-treated CD11b+Gr-1+ cells protected mice from post-influenza pneumococcal pneumonia. Further analysis revealed that the CAM-induced CD11b+Gr-1+ cell expansion was dependent on STAT3-mediated Bv8 production and may be facilitated by the presence of gut commensal microbiota. Lastly, an analysis of peripheral blood obtained from healthy volunteers following oral CAM administration showed a trend toward the expansion of human MDSC-like cells (Lineage-HLA-DR-CD11b+CD33+) with increased arginase 1 mRNA expression. Thus, CAM promoted the expansion of a unique population of immunosuppressive CD11b+Gr-1+ cells essential for the immunomodulatory properties of macrolides.


Assuntos
Claritromicina/farmacologia , Hormônios Gastrointestinais/metabolismo , Neuropeptídeos/metabolismo , Infecções por Orthomyxoviridae/complicações , Pneumonia Pneumocócica/tratamento farmacológico , Fator de Transcrição STAT3/metabolismo , Choque Séptico/tratamento farmacológico , Streptococcus pneumoniae/efeitos dos fármacos , Animais , Antibacterianos/farmacologia , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Hormônios Gastrointestinais/genética , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/microbiologia , Células Mieloides/virologia , Neuropeptídeos/genética , Orthomyxoviridae/patogenicidade , Infecções por Orthomyxoviridae/virologia , Fagocitose/efeitos dos fármacos , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/virologia , Fator de Transcrição STAT3/genética , Choque Séptico/induzido quimicamente
20.
Nature ; 499(7456): 97-101, 2013 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-23803760

RESUMO

Obesity has become more prevalent in most developed countries over the past few decades, and is increasingly recognized as a major risk factor for several common types of cancer. As the worldwide obesity epidemic has shown no signs of abating, better understanding of the mechanisms underlying obesity-associated cancer is urgently needed. Although several events were proposed to be involved in obesity-associated cancer, the exact molecular mechanisms that integrate these events have remained largely unclear. Here we show that senescence-associated secretory phenotype (SASP) has crucial roles in promoting obesity-associated hepatocellular carcinoma (HCC) development in mice. Dietary or genetic obesity induces alterations of gut microbiota, thereby increasing the levels of deoxycholic acid (DCA), a gut bacterial metabolite known to cause DNA damage. The enterohepatic circulation of DCA provokes SASP phenotype in hepatic stellate cells (HSCs), which in turn secretes various inflammatory and tumour-promoting factors in the liver, thus facilitating HCC development in mice after exposure to chemical carcinogen. Notably, blocking DCA production or reducing gut bacteria efficiently prevents HCC development in obese mice. Similar results were also observed in mice lacking an SASP inducer or depleted of senescent HSCs, indicating that the DCA-SASP axis in HSCs has key roles in obesity-associated HCC development. Moreover, signs of SASP were also observed in the HSCs in the area of HCC arising in patients with non-alcoholic steatohepatitis, indicating that a similar pathway may contribute to at least certain aspects of obesity-associated HCC development in humans as well. These findings provide valuable new insights into the development of obesity-associated cancer and open up new possibilities for its control.


Assuntos
Senescência Celular , Ácido Desoxicólico/metabolismo , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Células Estreladas do Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Obesidade/metabolismo , Animais , Antibacterianos/farmacologia , Bactérias/metabolismo , Ácidos e Sais Biliares/metabolismo , Carcinoma Hepatocelular/complicações , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/prevenção & controle , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Citocinas/metabolismo , Dano ao DNA/efeitos dos fármacos , Ácido Desoxicólico/sangue , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Modelos Animais de Doenças , Fígado Gorduroso/complicações , Fígado Gorduroso/patologia , Trato Gastrointestinal/efeitos dos fármacos , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/efeitos dos fármacos , Humanos , Interleucina-1beta/deficiência , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica , Obesidade/induzido quimicamente , Fenótipo , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA