Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 19(10): e1011691, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37847677

RESUMO

Even though gammaherpesvirus and parasitic infections are endemic in parts of the world, there is a lack of understanding about the outcome of coinfection. In humans, coinfections usually occur sequentially, with fluctuating order and timing in different hosts. However, experimental studies in mice generally do not address the variables of order and timing of coinfections. We sought to examine the variable of coinfection order in a system of gammaherpesvirus-helminth coinfection. Our previous work demonstrated that infection with the intestinal parasite, Heligmosomoides polygyrus, induced transient reactivation from latency of murine gammaherpesvirus-68 (MHV68). In this report, we reverse the order of coinfection, infecting with H. polygyrus first, followed by MHV68, and examined the effects of preexisting parasite infection on MHV68 acute and latent infection. We found that preexisting parasite infection increased the propensity of MHV68 to reactivate from latency. However, when we examined the mechanism for reactivation, we found that preexisting parasite infection increased the ability of MHV68 to reactivate in a vitamin A dependent manner, a distinct mechanism to what we found previously with parasite-induced reactivation after latency establishment. We determined that H. polygyrus infection increased both acute and latent MHV68 infection in a population of tissue resident macrophages, called large peritoneal macrophages. We demonstrate that this population of macrophages and vitamin A are required for increased acute and latent infection during parasite coinfection.


Assuntos
Coinfecção , Gammaherpesvirinae , Helmintos , Infecções por Herpesviridae , Infecção Latente , Doenças Parasitárias , Humanos , Animais , Camundongos , Ativação Viral , Latência Viral/fisiologia , Vitamina A , Linfócitos B , Infecções por Herpesviridae/complicações , Gammaherpesvirinae/fisiologia , Macrófagos , Camundongos Endogâmicos C57BL
2.
Elife ; 122023 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-37159507

RESUMO

Peristaltic movement of the intestine propels food down the length of the gastrointestinal tract to promote nutrient absorption. Interactions between intestinal macrophages and the enteric nervous system regulate gastrointestinal motility, yet we have an incomplete understanding of the molecular mediators of this crosstalk. Here, we identify complement component 1q (C1q) as a macrophage product that regulates gut motility. Macrophages were the predominant source of C1q in the mouse intestine and most extraintestinal tissues. Although C1q mediates the complement-mediated killing of bacteria in the bloodstream, we found that C1q was not essential for the immune defense of the intestine. Instead, C1q-expressing macrophages were located in the intestinal submucosal and myenteric plexuses where they were closely associated with enteric neurons and expressed surface markers characteristic of nerve-adjacent macrophages in other tissues. Mice with a macrophage-specific deletion of C1qa showed changes in enteric neuronal gene expression, increased neurogenic activity of peristalsis, and accelerated intestinal transit. Our findings identify C1q as a key regulator of gastrointestinal motility and provide enhanced insight into the crosstalk between macrophages and the enteric nervous system.


Assuntos
Complemento C1q , Sistema Nervoso Entérico , Camundongos , Animais , Complemento C1q/metabolismo , Motilidade Gastrointestinal/fisiologia , Macrófagos/metabolismo , Trato Gastrointestinal
3.
Sci Immunol ; 8(81): eabo2003, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36867675

RESUMO

Gut microbiota, specifically gut bacteria, are critical for effective immune checkpoint blockade therapy (ICT) for cancer. The mechanisms by which gut microbiota augment extraintestinal anticancer immune responses, however, are largely unknown. Here, we find that ICT induces the translocation of specific endogenous gut bacteria into secondary lymphoid organs and subcutaneous melanoma tumors. Mechanistically, ICT induces lymph node remodeling and dendritic cell (DC) activation, which facilitates the translocation of a selective subset of gut bacteria to extraintestinal tissues to promote optimal antitumor T cell responses in both the tumor-draining lymph nodes (TDLNs) and the primary tumor. Antibiotic treatment results in decreased gut microbiota translocation into mesenteric lymph nodes (MLNs) and TDLNs, diminished DC and effector CD8+ T cell responses, and attenuated responses to ICT. Our findings illuminate a key mechanism by which gut microbiota promote extraintestinal anticancer immunity.


Assuntos
Microbioma Gastrointestinal , Melanoma , Humanos , Inibidores de Checkpoint Imunológico , Linfócitos T CD8-Positivos , Linfonodos
4.
Nat Immunol ; 24(3): 531-544, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36658240

RESUMO

Immunoglobulin A (IgA) secretion by plasma cells, terminally differentiated B cells residing in the intestinal lamina propria, assures microbiome homeostasis and protects the host against enteric infections. Exposure to diet-derived and commensal-derived signals provides immune cells with organizing cues that instruct their effector function and dynamically shape intestinal immune responses at the mucosal barrier. Recent data have described metabolic and microbial inputs controlling T cell and innate lymphoid cell activation in the gut; however, whether IgA-secreting lamina propria plasma cells are tuned by local stimuli is completely unknown. Although antibody secretion is considered to be imprinted during B cell differentiation and therefore largely unaffected by environmental changes, a rapid modulation of IgA levels in response to intestinal fluctuations might be beneficial to the host. In the present study, we showed that dietary cholesterol absorption and commensal recognition by duodenal intestinal epithelial cells lead to the production of oxysterols, evolutionarily conserved lipids with immunomodulatory functions. Using conditional cholesterol 25-hydroxylase deleter mouse line we demonstrated that 7α,25-dihydroxycholesterol from epithelial cells is critical to restrain IgA secretion against commensal- and pathogen-derived antigens in the gut. Intestinal plasma cells sense oxysterols via the chemoattractant receptor GPR183 and couple their tissue positioning with IgA secretion. Our findings revealed a new mechanism linking dietary cholesterol and humoral immune responses centered around plasma cell localization for efficient mucosal protection.


Assuntos
Imunidade Inata , Plasmócitos , Animais , Camundongos , Colesterol na Dieta , Células Epiteliais , Imunoglobulina A , Mucosa Intestinal , Receptores Acoplados a Proteínas G , Intestinos
5.
J Biol Chem ; 298(2): 101463, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34864058

RESUMO

Interleukin (IL)-22 is a cytokine that plays a critical role in intestinal epithelial homeostasis. Its downstream functions are mediated through interaction with the heterodimeric IL-22 receptor and subsequent activation of signal transducer and activator of transcription 3 (STAT3). IL-22 signaling can induce transcription of genes necessary for intestinal epithelial cell proliferation, tissue regeneration, tight junction fortification, and antimicrobial production. Recent studies have also implicated IL-22 signaling in the regulation of intestinal epithelial fucosylation in mice. However, whether IL-22 regulates intestinal fucosylation in human intestinal epithelial cells and the molecular mechanisms that govern this process are unknown. Here, in experiments performed in human cell lines and human-derived enteroids, we show that IL-22 signaling regulates expression of the B3GNT7 transcript, which encodes a ß1-3-N-acetylglucosaminyltransferase that can participate in the synthesis of poly-N-acetyllactosamine (polyLacNAc) chains. Additionally, we find that IL-22 signaling regulates levels of the α1-3-fucosylated Lewis X (Lex) blood group antigen, and that this glycan epitope is primarily displayed on O-glycosylated intestinal epithelial glycoproteins. Moreover, we show that increased expression of B3GNT7 alone is sufficient to promote increased display of Lex-decorated carbohydrate glycan structures primarily on O-glycosylated intestinal epithelial glycoproteins. Together, these data identify B3GNT7 as an intermediary in IL-22-dependent induction of fucosylation of glycoproteins and uncover a novel role for B3GNT7 in intestinal glycosylation.


Assuntos
Células Epiteliais , Glicoproteínas , Interleucinas , Mucosa Intestinal , N-Acetilglucosaminiltransferases , Células Epiteliais/metabolismo , Glicoproteínas/metabolismo , Glicosilação , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , N-Acetilglucosaminiltransferases/biossíntese , N-Acetilglucosaminiltransferases/metabolismo , Polissacarídeos/metabolismo , Interleucina 22
6.
Ann N Y Acad Sci ; 1506(1): 55-73, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34414571

RESUMO

There is an increasing appreciation for the role of metabolism in cell signaling and cell decision making. Precise metabolic control is essential in development, as evident by the disorders caused by mutations in metabolic enzymes. The metabolic profile of cells is often cell-type specific, changing as cells differentiate or during tumorigenesis. Recent evidence has shown that changes in metabolism are not merely a consequence of changes in cell state but that metabolites can serve to promote and/or inhibit these changes. Metabolites can link metabolic pathways with cell signaling pathways via several mechanisms, for example, by serving as substrates for protein post-translational modifications, by affecting enzyme activity via allosteric mechanisms, or by altering epigenetic markers. Unraveling the complex interactions governing metabolism, gene expression, and protein activity that ultimately govern a cell's fate will require new tools and interactions across disciplines. On March 24 and 25, 2021, experts in cell metabolism, developmental biology, and human disease met virtually for the Keystone eSymposium, "Metabolic Decisions in Development and Disease." The discussions explored how metabolites impact cellular and developmental decisions in a diverse range of model systems used to investigate normal development, developmental disorders, dietary effects, and cancer-mediated changes in metabolism.


Assuntos
Congressos como Assunto/tendências , Desenvolvimento Humano/fisiologia , Doenças Metabólicas/fisiopatologia , Redes e Vias Metabólicas/fisiologia , Neoplasias/fisiopatologia , Relatório de Pesquisa , Animais , Epigênese Genética/fisiologia , Humanos , Doenças Metabólicas/genética , Neoplasias/genética , Transdução de Sinais/fisiologia
7.
Cell ; 184(16): 4154-4167.e12, 2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34324837

RESUMO

Environmental light cycles entrain circadian feeding behaviors in animals that produce rhythms in exposure to foodborne bacteria. Here, we show that the intestinal microbiota generates diurnal rhythms in innate immunity that synchronize with feeding rhythms to anticipate microbial exposure. Rhythmic expression of antimicrobial proteins was driven by daily rhythms in epithelial attachment by segmented filamentous bacteria (SFB), members of the mouse intestinal microbiota. Rhythmic SFB attachment was driven by the circadian clock through control of feeding rhythms. Mechanistically, rhythmic SFB attachment activated an immunological circuit involving group 3 innate lymphoid cells. This circuit triggered oscillations in epithelial STAT3 expression and activation that produced rhythmic antimicrobial protein expression and caused resistance to Salmonella Typhimurium infection to vary across the day-night cycle. Thus, host feeding rhythms synchronize with the microbiota to promote rhythms in intestinal innate immunity that anticipate exogenous microbial exposure.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Microbioma Gastrointestinal , Imunidade Inata , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Aderência Bacteriana , Adesão Celular , Células Epiteliais/microbiologia , Comportamento Alimentar , Intestino Delgado/microbiologia , Intestino Delgado/ultraestrutura , Linfócitos/metabolismo , Camundongos Endogâmicos C57BL , Muramidase/metabolismo , Proteínas Associadas a Pancreatite/metabolismo , Fator de Transcrição STAT3/metabolismo , Salmonelose Animal/microbiologia , Transdução de Sinais
8.
Science ; 365(6460): 1428-1434, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31604271

RESUMO

Circadian rhythmicity is a defining feature of mammalian metabolism that synchronizes metabolic processes to day-night light cycles. Here, we show that the intestinal microbiota programs diurnal metabolic rhythms in the mouse small intestine through histone deacetylase 3 (HDAC3). The microbiota induced expression of intestinal epithelial HDAC3, which was recruited rhythmically to chromatin, and produced synchronized diurnal oscillations in histone acetylation, metabolic gene expression, and nutrient uptake. HDAC3 also functioned noncanonically to coactivate estrogen-related receptor α, inducing microbiota-dependent rhythmic transcription of the lipid transporter gene Cd36 and promoting lipid absorption and diet-induced obesity. Our findings reveal that HDAC3 integrates microbial and circadian cues for regulation of diurnal metabolic rhythms and pinpoint a key mechanism by which the microbiota controls host metabolism.


Assuntos
Ritmo Circadiano , Células Epiteliais/metabolismo , Microbioma Gastrointestinal , Histona Desacetilases/metabolismo , Intestino Delgado/metabolismo , Acetilação , Animais , Antígenos CD36/metabolismo , Cromatina/metabolismo , Colo , Dieta Hiperlipídica , Vida Livre de Germes , Intestino Delgado/citologia , Síndrome do Jet Lag , Metabolismo dos Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Receptores de Estrogênio/metabolismo , Receptor ERRalfa Relacionado ao Estrogênio
9.
Cell Host Microbe ; 25(6): 777-788.e8, 2019 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-31101494

RESUMO

Vitamin A deficiency increases susceptibility to skin infection. However, the mechanisms by which vitamin A regulates skin immunity remain unclear. Here, we show that resistin-like molecule α (RELMα), a small secreted cysteine-rich protein, is expressed by epidermal keratinocytes and sebocytes and serves as an antimicrobial protein that is required for vitamin-A-dependent resistance to skin infection. RELMα was induced by microbiota colonization of the murine skin, was bactericidal in vitro, and was protected against bacterial infection of the skin in vivo. RELMα expression required dietary vitamin A and was induced by the therapeutic vitamin A analog isotretinoin, which protected against skin infection in a RELMα-dependent manner. The RELM family member Resistin was expressed in human skin, was induced by vitamin A analogs, and killed skin bacteria, indicating a conserved function for RELM proteins in skin innate immunity. Our findings provide insight into how vitamin A promotes resistance to skin infection.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Fatores Imunológicos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Dermatopatias Bacterianas/prevenção & controle , Pele/imunologia , Vitamina A/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Humanos , Camundongos , Resistina/metabolismo , Dermatopatias Bacterianas/imunologia , Ativação Transcricional/efeitos dos fármacos
10.
Proc Natl Acad Sci U S A ; 116(22): 10911-10916, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31097581

RESUMO

Vitamin A is a dietary component that is essential for the development of intestinal immunity. Vitamin A is absorbed and converted to its bioactive derivatives retinol and retinoic acid by the intestinal epithelium, yet little is known about how epithelial cells regulate vitamin A-dependent intestinal immunity. Here we show that epithelial cell expression of the transcription factor retinoic acid receptor ß (RARß) is essential for vitamin A-dependent intestinal immunity. Epithelial RARß activated vitamin A-dependent expression of serum amyloid A (SAA) proteins by binding directly to Saa promoters. In accordance with the known role of SAAs in regulating Th17 cell effector function, epithelial RARß promoted IL-17 production by intestinal Th17 cells. More broadly, epithelial RARß was required for the development of key vitamin A-dependent adaptive immune responses, including CD4+ T-cell homing to the intestine and the development of IgA-producing intestinal B cells. Our findings provide insight into how the intestinal epithelium senses dietary vitamin A status to regulate adaptive immunity, and highlight the role of epithelial cells in regulating intestinal immunity in response to diet.


Assuntos
Imunidade nas Mucosas/fisiologia , Mucosa Intestinal/metabolismo , Receptores do Ácido Retinoico/metabolismo , Proteína Amiloide A Sérica/metabolismo , Vitamina A/metabolismo , Animais , Linhagem Celular , Microbioma Gastrointestinal/fisiologia , Células Hep G2 , Humanos , Camundongos , Receptores do Ácido Retinoico/genética , Proteína Amiloide A Sérica/genética
11.
Brain Behav Immun ; 74: 176-185, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30195028

RESUMO

The mammalian nervous system communicates important information about the environment to the immune system, but the underlying mechanisms are largely unknown. Secondary lymphoid organs are highly innervated by sympathetic neurons that secrete norepinephrine (NE) as the primary neurotransmitter. Immune cells express adrenergic receptors, enabling the sympathetic nervous system to directly control immune function. NE is a potent immunosuppressive factor and markedly inhibits TNF-α secretion from innate cells in response to lipopolysaccharide (LPS). In this study, we demonstrate that NE blocks the secretion of a variety of proinflammatory cytokines by rapidly inducing IL-10 secretion from innate cells in response to multiple Toll-like receptor (TLR) signals. NE mediated these effects exclusively through the ß2-adrenergic receptor (ADRB2). Consequently, Adrb2-/- animals were more susceptible to L. monocytogenes infection and to intestinal inflammation in a dextran sodium sulfate (DSS) model of colitis. Further, Adrb2-/- animals rapidly succumbed to endotoxemia in response to a sub-lethal LPS challenge and exhibited elevated serum levels of TNF-α and reduced IL-10. LPS-mediated lethality in WT animals was rescued by administering a ß 2-specific agonist and in Adrb2-/- animals by exogenous IL-10. These findings reveal a critical role for ADRB2 signaling in controlling inflammation through the rapid induction of IL-10. Our findings provide a fundamental insight into how the sympathetic nervous system controls a critical facet of immune function through ADRB2 signaling.


Assuntos
Interleucina-10/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Animais , Citocinas/metabolismo , Sistema Imunitário/efeitos dos fármacos , Inflamação/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Norepinefrina/metabolismo , Norepinefrina/farmacologia , Norepinefrina/fisiologia , Transdução de Sinais , Sistema Nervoso Simpático/efeitos dos fármacos , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
12.
J Clin Invest ; 128(11): 4970-4979, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30106382

RESUMO

Graft-versus-host disease (GVHD) in the gastrointestinal (GI) tract remains the major cause of morbidity and nonrelapse mortality after BM transplantation (BMT). The Paneth cell protein regenerating islet-derived 3α (REG3α) is a biomarker specific for GI GVHD. REG3α serum levels rose in the systematic circulation as GVHD progressively destroyed Paneth cells and reduced GI epithelial barrier function. Paradoxically, GVHD suppressed intestinal REG3γ (the mouse homolog of human REG3α), and the absence of REG3γ in BMT recipients intensified GVHD but did not change the composition of the microbiome. IL-22 administration restored REG3γ production and prevented apoptosis of both intestinal stem cells (ISCs) and Paneth cells, but this protection was completely abrogated in Reg3g-/- mice. In vitro, addition of REG3α reduced the apoptosis of colonic cell lines. Strategies that increase intestinal REG3α/γ to promote crypt regeneration may offer a novel, nonimmunosuppressive approach for GVHD and perhaps for other diseases involving the ISC niche, such as inflammatory bowel disease.


Assuntos
Apoptose , Transplante de Medula Óssea , Colo/metabolismo , Doença Enxerto-Hospedeiro/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Proteínas Associadas a Pancreatite/metabolismo , Celulas de Paneth/metabolismo , Transdução de Sinais , Animais , Sobrevivência Celular/genética , Colo/patologia , Feminino , Doença Enxerto-Hospedeiro/patologia , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Associadas a Pancreatite/genética , Celulas de Paneth/patologia , Estudos Prospectivos , Transplante Homólogo
13.
Autophagy ; 14(4): 719-721, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29388875

RESUMO

Secretion of antimicrobial proteins is an important host defense mechanism against bacteria, yet how secretory cells maintain function during bacterial invasion has been unclear. We discovered that Paneth cells, specialized secretory cells in the small intestine, react to bacterial invasion by rerouting a critical secreted antibacterial protein through a macroautophagy/autophagy-based secretion system termed secretory autophagy. Mice harboring a mutation in an essential autophagy gene, a mutation which is common in Crohn disease patients, cannot reroute their antimicrobial cargo during bacterial invasion and thus have compromised innate immunity. We showed that this alternative secretion system is triggered by both a cell-intrinsic mechanism, involving the ER stress response, and a cell-extrinsic mechanism, involving subepithelial innate immune cells. Our findings uncover a new role for secretory autophagy in host defense and suggest how a mutation in an autophagy gene can predispose individuals to Crohn disease.


Assuntos
Autofagia/fisiologia , Estresse do Retículo Endoplasmático/genética , Intestino Delgado/microbiologia , Celulas de Paneth/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Transporte Biológico/fisiologia , Proteínas de Transporte/metabolismo , Doença de Crohn/genética , Doença de Crohn/microbiologia , Muramidase/metabolismo , Mutação/genética , Celulas de Paneth/microbiologia
14.
Cell Host Microbe ; 23(2): 177-190.e4, 2018 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-29358083

RESUMO

The protozoan parasite Toxoplasma gondii triggers severe small intestinal immunopathology characterized by IFN-γ- and intestinal microbiota-mediated inflammation, Paneth cell loss, and bacterial dysbiosis. Paneth cells are a prominent secretory epithelial cell type that resides at the base of intestinal crypts and releases antimicrobial peptides. We demonstrate that the microbiota triggers basal Paneth cell-specific autophagy via induction of IFN-γ, a known trigger of autophagy, to maintain intestinal homeostasis. Deletion of the autophagy protein Atg5 specifically in Paneth cells results in exaggerated intestinal inflammation characterized by complete destruction of the intestinal crypts resembling that seen in pan-epithelial Atg5-deficient mice. Additionally, lack of functional autophagy in Paneth cells within intestinal organoids and T. gondii-infected mice causes increased sensitivity to the proinflammatory cytokine TNF along with increased intestinal permeability, leading to exaggerated microbiota- and IFN-γ-dependent intestinal immunopathology. Thus, Atg5 expression in Paneth cells is essential for tissue protection against cytokine-mediated immunopathology during acute gastrointestinal infection.


Assuntos
Proteína 5 Relacionada à Autofagia/metabolismo , Autofagia/imunologia , Interferon gama/imunologia , Celulas de Paneth/imunologia , Toxoplasma/imunologia , Toxoplasmose Animal/patologia , Animais , Proteína 5 Relacionada à Autofagia/genética , Linfócitos T CD4-Positivos/imunologia , Disbiose/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Toxoplasmose Animal/imunologia , Toxoplasmose Animal/parasitologia , Fator de Necrose Tumoral alfa/imunologia
15.
Proc Natl Acad Sci U S A ; 114(42): 11027-11033, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-28973871

RESUMO

The mammalian intestine is colonized by trillions of bacteria that perform essential metabolic functions for their hosts. The mutualistic nature of this relationship depends on maintaining spatial segregation between these bacteria and the intestinal epithelial surface. This segregation is achieved in part by the presence of a dense mucus layer at the epithelial surface and by the production of antimicrobial proteins that are secreted by epithelial cells into the mucus layer. Here, we show that resistin-like molecule ß (RELMß) is a bactericidal protein that limits contact between Gram-negative bacteria and the colonic epithelial surface. Mouse and human RELMß selectively killed Gram-negative bacteria by forming size-selective pores that permeabilized bacterial membranes. In mice lacking RELMß, Proteobacteria were present in the inner mucus layer and invaded mucosal tissues. Another RELM family member, human resistin, was also bactericidal, suggesting that bactericidal activity is a conserved function of the RELM family. Our findings thus identify the RELM family as a unique family of bactericidal proteins and show that RELMß promotes host-bacterial mutualism by regulating the spatial segregation between the microbiota and the intestinal epithelium.


Assuntos
Microbioma Gastrointestinal , Bactérias Gram-Negativas , Hormônios Ectópicos/fisiologia , Mucosa Intestinal/microbiologia , Animais , Humanos , Imunidade Inata , Peptídeos e Proteínas de Sinalização Intercelular , Mucosa Intestinal/imunologia , Metabolismo dos Lipídeos , Camundongos , Resistina/fisiologia , Simbiose
16.
Science ; 357(6355): 1047-1052, 2017 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-28751470

RESUMO

Intestinal Paneth cells limit bacterial invasion by secreting antimicrobial proteins, including lysozyme. However, invasive pathogens can disrupt the Golgi apparatus, interfering with secretion and compromising intestinal antimicrobial defense. Here we show that during bacterial infection, lysozyme is rerouted via secretory autophagy, an autophagy-based alternative secretion pathway. Secretory autophagy was triggered in Paneth cells by bacteria-induced endoplasmic reticulum (ER) stress, required extrinsic signals from innate lymphoid cells, and limited bacterial dissemination. Secretory autophagy was disrupted in Paneth cells of mice harboring a mutation in autophagy gene Atg16L1 that confers increased risk for Crohn's disease in humans. Our findings identify a role for secretory autophagy in intestinal defense and suggest why Crohn's disease is associated with genetic mutations that affect both the ER stress response and autophagy.


Assuntos
Estresse do Retículo Endoplasmático/imunologia , Muramidase/metabolismo , Celulas de Paneth/imunologia , Celulas de Paneth/metabolismo , Infecções por Salmonella/imunologia , Salmonella enterica , Animais , Autofagia/genética , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Doença de Crohn/genética , Doença de Crohn/imunologia , Doença de Crohn/microbiologia , Estresse do Retículo Endoplasmático/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Celulas de Paneth/enzimologia , Infecções por Salmonella/genética
17.
Cell Rep ; 19(13): 2756-2770, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28658623

RESUMO

Although NOD2 is the major inflammatory bowel disease susceptibility gene, its role in colorectal tumorigenesis is poorly defined. Here, we show that Nod2-deficient mice are highly susceptible to experimental colorectal tumorigenesis independent of gut microbial dysbiosis. Interestingly, the expression of inflammatory genes and the activation of inflammatory pathways, including NF-κB, ERK, and STAT3 are significantly higher in Nod2-/- mouse colons during colitis and colorectal tumorigenesis, but not at homeostasis. Consistent with higher inflammation, there is greater proliferation of epithelial cells in hyperplastic regions of Nod2-/- colons. In vitro studies demonstrate that, while NOD2 activates the NF-κB and MAPK pathways in response to MDP, it inhibits TLR-mediated activation of NF-κB and MAPK. Notably, NOD2-mediated downregulation of NF-κB and MAPK is associated with the induction of IRF4. Taken together, NOD2 plays a critical role in the suppression of inflammation and tumorigenesis in the colon via downregulation of the TLR signaling pathways.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Proteína Adaptadora de Sinalização NOD2/genética , Proteína Adaptadora de Sinalização NOD2/metabolismo , Receptores Toll-Like/metabolismo , Animais , Carcinogênese , Regulação para Baixo , Feminino , Fatores Reguladores de Interferon/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Transdução de Sinais , Receptores Toll-Like/genética
18.
Proc Natl Acad Sci U S A ; 114(7): E1196-E1204, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28137874

RESUMO

Class-switch recombination (CSR) alters the Ig isotype to diversify antibody effector functions. IgD CSR is a rare event, and its regulation is poorly understood. We report that deficiency of 53BP1, a DNA damage-response protein, caused age-dependent overproduction of secreted IgD resulting from increased IgD CSR exclusively within B cells of mucosa-associated lymphoid tissues. IgD overproduction was dependent on activation-induced cytidine deaminase, hematopoietic MyD88 expression, and an intact microbiome, against which circulating IgD, but not IgM, was reactive. IgD CSR occurred via both alternative nonhomologous end-joining and homologous recombination pathways. Microbiota-dependent IgD CSR also was detected in nasal-associated lymphoid tissue of WT mice. These results identify a pathway, present in WT mice and hyperactivated in 53BP1-deficient mice, by which microbiota signal via Toll-like receptors to elicit IgD CSR.


Assuntos
Switching de Imunoglobulina , Imunoglobulina D/imunologia , Tecido Linfoide/imunologia , Microbiota/imunologia , Mucosa/imunologia , Animais , Citidina Desaminase/genética , Citidina Desaminase/imunologia , Citidina Desaminase/metabolismo , Reparo do DNA por Junção de Extremidades , Feminino , Imunoglobulina D/genética , Imunoglobulina D/metabolismo , Tecido Linfoide/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microbiota/genética , Mucosa/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Recombinação Genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/deficiência , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/imunologia
20.
Adv Immunol ; 126: 129-72, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25727289

RESUMO

The epithelial surfaces of the mammalian intestine interface directly with the external environment and thus continuously encounter pathogenic bacteria, fungi, viruses, and parasites. The intestinal epithelium is also closely associated with complex communities of symbiotic microorganisms. Intestinal epithelial cells are thus faced with the unique challenge of directly interacting with enormous numbers of microbes that include both pathogens and symbionts. As a result, gut epithelia have evolved an array of strategies that contribute to host immunity. This chapter considers the various mechanisms used by epithelial cells to limit microbial invasion of host tissues, shape the composition of indigenous microbial communities, and coordinate the adaptive immune response to microorganisms. Study of intestinal epithelial cells has contributed fundamental insights into intestinal immune homeostasis and has revealed how impaired epithelial cell function can contribute to inflammatory disease.


Assuntos
Células Epiteliais/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Microbiota , Imunidade Adaptativa , Animais , Peptídeos Catiônicos Antimicrobianos/imunologia , Autofagia , Homeostase/imunologia , Humanos , Imunidade Inata , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/citologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA