Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Immunol ; 36: 193-220, 2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29328787

RESUMO

Retroviruses are genome invaders that have shared a long history of coevolution with vertebrates and their immune system. Found endogenously in genomes as traces of past invasions, retroviruses are also considerable threats to human health when they exist as exogenous viruses such as HIV. The immune response to retroviruses is engaged by germline-encoded sensors of innate immunity that recognize viral components and damage induced by the infection. This response develops with the induction of antiviral effectors and launching of the clonal adaptive immune response, which can contribute to protective immunity. However, retroviruses efficiently evade the immune response, owing to their rapid evolution. The failure of specialized immune cells to respond, a form of neglect, may also contribute to inadequate antiretroviral immune responses. Here, we discuss the mechanisms by which immune responses to retroviruses are mounted at the molecular, cellular, and organismal levels. We also discuss how intrinsic, innate, and adaptive immunity may cooperate or conflict during the generation of immune responses.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Retroviridae/imunologia , Imunidade Adaptativa , Animais , Humanos , Evasão da Resposta Imune , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Imunidade Inata , Infecções por Retroviridae/metabolismo
2.
Nat Immunol ; 25(7): 1193-1206, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38834865

RESUMO

Immune cells experience large cell shape changes during environmental patrolling because of the physical constraints that they encounter while migrating through tissues. These cells can adapt to such deformation events using dedicated shape-sensing pathways. However, how shape sensing affects immune cell function is mostly unknown. Here, we identify a shape-sensing mechanism that increases the expression of the chemokine receptor CCR7 and guides dendritic cell migration from peripheral tissues to lymph nodes at steady state. This mechanism relies on the lipid metabolism enzyme cPLA2, requires nuclear envelope tensioning and is finely tuned by the ARP2/3 actin nucleation complex. We also show that this shape-sensing axis reprograms dendritic cell transcription by activating an IKKß-NF-κB-dependent pathway known to control their tolerogenic potential. These results indicate that cell shape changes experienced by immune cells can define their migratory behavior and immunoregulatory properties and reveal a contribution of the physical properties of tissues to adaptive immunity.


Assuntos
Movimento Celular , Células Dendríticas , Homeostase , Linfonodos , Camundongos Endogâmicos C57BL , Receptores CCR7 , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Linfonodos/imunologia , Linfonodos/citologia , Receptores CCR7/metabolismo , Camundongos , Movimento Celular/imunologia , Forma Celular , NF-kappa B/metabolismo , Camundongos Knockout , Transdução de Sinais/imunologia , Quinase I-kappa B/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo
3.
Cell ; 184(20): 5230-5246.e22, 2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34551315

RESUMO

Although mutations leading to a compromised nuclear envelope cause diseases such as muscular dystrophies or accelerated aging, the consequences of mechanically induced nuclear envelope ruptures are less known. Here, we show that nuclear envelope ruptures induce DNA damage that promotes senescence in non-transformed cells and induces an invasive phenotype in human breast cancer cells. We find that the endoplasmic reticulum (ER)-associated exonuclease TREX1 translocates into the nucleus after nuclear envelope rupture and is required to induce DNA damage. Inside the mammary duct, cellular crowding leads to nuclear envelope ruptures that generate TREX1-dependent DNA damage, thereby driving the progression of in situ carcinoma to the invasive stage. DNA damage and nuclear envelope rupture markers were also enriched at the invasive edge of human tumors. We propose that DNA damage in mechanically challenged nuclei could affect the pathophysiology of crowded tissues by modulating proliferation and extracellular matrix degradation of normal and transformed cells.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Dano ao DNA , Exodesoxirribonucleases/metabolismo , Membrana Nuclear/metabolismo , Fosfoproteínas/metabolismo , Animais , Linhagem Celular , Senescência Celular , Colágeno/metabolismo , Progressão da Doença , Feminino , Humanos , Camundongos , Invasividade Neoplásica , Membrana Nuclear/ultraestrutura , Proteólise , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cell ; 175(2): 488-501.e22, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30270045

RESUMO

Detection of viruses by innate immune sensors induces protective antiviral immunity. The viral DNA sensor cyclic GMP-AMP synthase (cGAS) is necessary for detection of HIV by human dendritic cells and macrophages. However, synthesis of HIV DNA during infection is not sufficient for immune activation. The capsid protein, which associates with viral DNA, has a pivotal role in enabling cGAS-mediated immune activation. We now find that NONO is an essential sensor of the HIV capsid in the nucleus. NONO protein directly binds capsid with higher affinity for weakly pathogenic HIV-2 than highly pathogenic HIV-1. Upon infection, NONO is essential for cGAS activation by HIV and cGAS association with HIV DNA in the nucleus. NONO recognizes a conserved region in HIV capsid with limited tolerance for escape mutations. Detection of nuclear viral capsid by NONO to promote DNA sensing by cGAS reveals an innate strategy to achieve distinction of viruses from self in the nucleus.


Assuntos
Proteínas do Capsídeo/imunologia , Proteínas Associadas à Matriz Nuclear/imunologia , Proteínas Associadas à Matriz Nuclear/fisiologia , Fatores de Transcrição de Octâmero/imunologia , Fatores de Transcrição de Octâmero/fisiologia , Proteínas de Ligação a RNA/imunologia , Proteínas de Ligação a RNA/fisiologia , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/fisiologia , Núcleo Celular/metabolismo , DNA Viral/genética , DNA Viral/imunologia , Proteínas de Ligação a DNA , Células Dendríticas/imunologia , Infecções por HIV/imunologia , HIV-1/genética , HIV-1/imunologia , HIV-2/genética , HIV-2/imunologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata/imunologia , Macrófagos/imunologia , Proteínas de Membrana/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/fisiologia , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/imunologia
5.
Nature ; 617(7960): 386-394, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37100912

RESUMO

Inflammation is a complex physiological process triggered in response to harmful stimuli1. It involves cells of the immune system capable of clearing sources of injury and damaged tissues. Excessive inflammation can occur as a result of infection and is a hallmark of several diseases2-4. The molecular bases underlying inflammatory responses are not fully understood. Here we show that the cell surface glycoprotein CD44, which marks the acquisition of distinct cell phenotypes in the context of development, immunity and cancer progression, mediates the uptake of metals including copper. We identify a pool of chemically reactive copper(II) in mitochondria of inflammatory macrophages that catalyses NAD(H) redox cycling by activating hydrogen peroxide. Maintenance of NAD+ enables metabolic and epigenetic programming towards the inflammatory state. Targeting mitochondrial copper(II) with supformin (LCC-12), a rationally designed dimer of metformin, induces a reduction of the NAD(H) pool, leading to metabolic and epigenetic states that oppose macrophage activation. LCC-12 interferes with cell plasticity in other settings and reduces inflammation in mouse models of bacterial and viral infections. Our work highlights the central role of copper as a regulator of cell plasticity and unveils a therapeutic strategy based on metabolic reprogramming and the control of epigenetic cell states.


Assuntos
Plasticidade Celular , Cobre , Inflamação , Transdução de Sinais , Animais , Camundongos , Cobre/metabolismo , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , NAD/metabolismo , Transdução de Sinais/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Peróxido de Hidrogênio/metabolismo , Epigênese Genética/efeitos dos fármacos , Metformina/análogos & derivados , Oxirredução , Plasticidade Celular/efeitos dos fármacos , Plasticidade Celular/genética , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/genética
6.
Nat Immunol ; 17(2): 140-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26657003

RESUMO

Innate sensing of pathogens initiates inflammatory cytokine responses that need to be tightly controlled. We found here that after engagement of Toll-like receptors (TLRs) in myeloid cells, deficient sumoylation caused increased secretion of transcription factor NF-κB-dependent inflammatory cytokines and a massive type I interferon signature. In mice, diminished sumoylation conferred susceptibility to endotoxin shock and resistance to viral infection. Overproduction of several NF-κB-dependent inflammatory cytokines required expression of the type I interferon receptor, which identified type I interferon as a central sumoylation-controlled hub for inflammation. Mechanistically, the small ubiquitin-like modifier SUMO operated from a distal enhancer of the gene encoding interferon-ß (Ifnb1) to silence both basal and stimulus-induced activity of the Ifnb1 promoter. Therefore, sumoylation restrained inflammation by silencing Ifnb1 expression and by strictly suppressing an unanticipated priming by type I interferons of the TLR-induced production of inflammatory cytokines.


Assuntos
Resistência à Doença , Regulação da Expressão Gênica , Imunidade Inata , Imunomodulação , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Sumoilação , Animais , Cromatina/genética , Cromatina/metabolismo , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Elementos Facilitadores Genéticos , Perfilação da Expressão Gênica , Loci Gênicos , Inflamação/virologia , Mediadores da Inflamação/metabolismo , Interferon beta/metabolismo , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Knockout , Ligação Proteica , Receptor de Interferon alfa e beta/metabolismo , Elementos Reguladores de Transcrição , Proteína SUMO-1/metabolismo , Choque Séptico/genética , Choque Séptico/imunologia , Choque Séptico/metabolismo , Transdução de Sinais , Sumoilação/genética , Sumoilação/imunologia , Receptores Toll-Like/metabolismo
7.
Cell ; 147(2): 271-4, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-22000008

RESUMO

Two groups have identified SAMHD1, a protein encoded by an Aicardi-Goutières Syndrome susceptibility gene, as the factor that restricts infection of macrophages and dendritic cells with HIV-1. Here we discuss implications of this discovery for induction of antiviral protective immunity.


Assuntos
Infecções por HIV/imunologia , HIV-1 , Evasão da Resposta Imune , Imunidade Inata , Vacinas contra a AIDS/imunologia , Imunidade Adaptativa , Doenças Autoimunes do Sistema Nervoso/genética , Células Dendríticas/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/virologia , Malformações do Sistema Nervoso/genética
8.
Proc Natl Acad Sci U S A ; 119(17): e2107394119, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35439048

RESUMO

Tumor associated macrophages (TAMs), which differentiate from circulating monocytes, are pervasive across human cancers and comprise heterogeneous populations. The contribution of tumor-derived signals to TAM heterogeneity is not well understood. In particular, tumors release both soluble factors and extracellular vesicles (EVs), whose respective impact on TAM precursors may be different. Here, we show that triple negative breast cancer cells (TNBCs) release EVs and soluble molecules promoting monocyte differentiation toward distinct macrophage fates. EVs specifically promoted proinflammatory macrophages bearing an interferon response signature. The combination in TNBC EVs of surface CSF-1 promoting survival and cargoes promoting cGAS/STING or other activation pathways led to differentiation of this particular macrophage subset. Notably, macrophages expressing the EV-induced signature were found among patients' TAMs. Furthermore, higher expression of this signature was associated with T cell infiltration and extended patient survival. Together, this data indicates that TNBC-released CSF-1-bearing EVs promote a tumor immune microenvironment associated with a better prognosis in TNBC patients.


Assuntos
Vesículas Extracelulares , Neoplasias de Mama Triplo Negativas , Vesículas Extracelulares/fisiologia , Humanos , Macrófagos , Neoplasias de Mama Triplo Negativas/patologia
9.
EMBO J ; 39(20): e105505, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32945564

RESUMO

Centromeres are built on repetitive DNA sequences (CenDNA) and a specific chromatin enriched with the histone H3 variant CENP-A, the epigenetic mark that identifies centromere position. Here, we interrogate the importance of CenDNA in centromere specification by developing a system to rapidly remove and reactivate CENP-A (CENP-AOFF/ON ). Using this system, we define the temporal cascade of events necessary to maintain centromere position. We unveil that CENP-B bound to CenDNA provides memory for maintenance on human centromeres by promoting de novo CENP-A deposition. Indeed, lack of CENP-B favors neocentromere formation under selective pressure. Occasionally, CENP-B triggers centromere re-activation initiated by CENP-C, but not CENP-A, recruitment at both ectopic and native centromeres. This is then sufficient to initiate the CENP-A-based epigenetic loop. Finally, we identify a population of CENP-A-negative, CENP-B/C-positive resting CD4+ T cells capable to re-express and reassembles CENP-A upon cell cycle entry, demonstrating the physiological importance of the genetic memory.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Proteína Centromérica A/metabolismo , Proteína B de Centrômero/metabolismo , Centrômero/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Segregação de Cromossomos , Nucleossomos/genética , Linfócitos T CD4-Positivos/citologia , Sistemas CRISPR-Cas , Ciclo Celular , Linhagem Celular Tumoral , Centrômero/genética , Segregação de Cromossomos/genética , Biologia Computacional , Epigênese Genética , Marcação de Genes , Humanos , Hibridização in Situ Fluorescente , Nucleossomos/metabolismo , RNA Interferente Pequeno
10.
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
11.
Cell ; 132(6): 1039-48, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18358815

RESUMO

Of all cells, human erythrocytes express the highest level of the Glut1 glucose transporter. However, the regulation and function of Glut1 during erythropoiesis are not known. Here, we report that glucose transport actually decreases during human erythropoiesis despite a >3-log increase in Glut1 transcripts. In contrast, Glut1-mediated transport of L-dehydroascorbic acid (DHA), an oxidized form of ascorbic acid (AA), is dramatically enhanced. We identified stomatin, an integral erythrocyte membrane protein, as regulating the switch from glucose to DHA transport. Notably though, we found that erythrocyte Glut1 and associated DHA uptake are unique traits of humans and the few other mammals that have lost the ability to synthesize AA from glucose. Accordingly, we show that mice, a species capable of synthesizing AA, express Glut4 but not Glut1 in mature erythrocytes. Thus, erythrocyte-specific coexpression of Glut1 with stomatin constitutes a compensatory mechanism in mammals that are unable to synthesize vitamin C.


Assuntos
Ácido Ascórbico/metabolismo , Ácido Desidroascórbico/metabolismo , Eritrócitos/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Regiões 5' não Traduzidas , Animais , Transporte Biológico , Linhagem Celular , Eritropoese , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 4/metabolismo , Humanos , Mamíferos , Proteínas de Membrana/metabolismo , Transfecção
12.
Immunity ; 39(6): 1132-42, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24269171

RESUMO

HIV-2 is less pathogenic for humans than HIV-1 and might provide partial cross-protection from HIV-1-induced pathology. Although both viruses replicate in the T cells of infected patients, only HIV-2 replicates efficiently in dendritic cells (DCs) and activates innate immune pathways. How HIV is sensed in DC is unknown. Capsid-mutated HIV-2 revealed that sensing by the host requires viral cDNA synthesis, but not nuclear entry or genome integration. The HIV-1 capsid prevented viral cDNA sensing up to integration, allowing the virus to escape innate recognition. In contrast, DCs sensed capsid-mutated HIV-1 and enhanced stimulation of T cells in the absence of productive infection. Finally, we found that DC sensing of HIV-1 and HIV-2 required the DNA sensor cGAS. Thus, the HIV capsid is a determinant of innate sensing of the viral cDNA by cGAS in dendritic cells. This pathway might potentially be harnessed to develop effective vaccines against HIV-1.


Assuntos
Capsídeo/imunologia , DNA Complementar/metabolismo , Células Dendríticas , Infecções por HIV/imunologia , HIV-1/imunologia , HIV-2/imunologia , Nucleotidiltransferases/metabolismo , Células Cultivadas , DNA Viral/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/virologia , Infecções por HIV/virologia , HIV-1/genética , HIV-1/metabolismo , HIV-2/genética , HIV-2/metabolismo , Humanos , Imunidade Inata/fisiologia , Modelos Biológicos
13.
Nat Immunol ; 9(6): 641-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18454151

RESUMO

T(H)-17 cells are interleukin 17 (IL-17)-secreting CD4+ T helper cells involved in autoimmune disease and mucosal immunity. In naive CD4+ T cells from mice, IL-17 is expressed in response to a combination of IL-6 or IL-21 and transforming growth factor-beta (TGF-beta) and requires induction of the nuclear receptor RORgammat. It has been suggested that the differentiation of human T(H)-17 cells is independent of TGF-beta and thus differs fundamentally from that in mice. We show here that TGF-beta, IL-1beta and IL-6, IL-21 or IL-23 in serum-free conditions were necessary and sufficient to induce IL-17 expression in naive human CD4+ T cells from cord blood. TGF-beta upregulated RORgammat expression but simultaneously inhibited its ability to induce IL-17 expression. Inflammatory cytokines relieved this inhibition and increased RORgammat-directed IL-17 expression. Other gene products detected in T(H)-17 cells after RORgammat induction included the chemokine receptor CCR6, the IL-23 receptor, IL-17F and IL-26. Our studies identify RORgammat as having a central function in the differentiation of human T(H)-17 cells from naive CD4+ T cells and suggest that similar cytokine pathways are involved in this process in mice and humans.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/imunologia , Interleucina-17/metabolismo , Receptores do Ácido Retinoico/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Receptores Citoplasmáticos e Nucleares/metabolismo
14.
Hepatology ; 68(5): 1695-1709, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29679386

RESUMO

Chronic hepatitis B virus (HBV) infection is a major cause of chronic liver disease and cancer worldwide. The mechanisms of viral genome sensing and the evasion of innate immune responses by HBV infection are still poorly understood. Recently, the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) was identified as a DNA sensor. In this study, we investigated the functional role of cGAS in sensing HBV infection and elucidate the mechanisms of viral evasion. We performed functional studies including loss-of-function and gain-of-function experiments combined with cGAS effector gene expression profiling in an infectious cell culture model, primary human hepatocytes, and HBV-infected human liver chimeric mice. Here, we show that cGAS is expressed in the human liver, primary human hepatocytes, and human liver chimeric mice. While naked relaxed-circular HBV DNA is sensed in a cGAS-dependent manner in hepatoma cell lines and primary human hepatocytes, host cell recognition of viral nucleic acids is abolished during HBV infection, suggesting escape from sensing, likely during packaging of the genome into the viral capsid. While the hepatocyte cGAS pathway is functionally active, as shown by reduction of viral covalently closed circular DNA levels in gain-of-function studies, HBV infection suppressed cGAS expression and function in cell culture models and humanized mice. Conclusion: HBV exploits multiple strategies to evade sensing and antiviral activity of cGAS and its effector pathways.


Assuntos
Vírus da Hepatite B/patogenicidade , Hepatite B/fisiopatologia , Hepatócitos/virologia , Evasão da Resposta Imune/fisiologia , Nucleotídeos Cíclicos/metabolismo , Animais , Western Blotting , Técnicas de Cultura de Células , DNA Viral/imunologia , Perfilação da Expressão Gênica/métodos , Hepatite B/imunologia , Hepatócitos/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune/imunologia , Hibridização in Situ Fluorescente/métodos , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
15.
PLoS Pathog ; 12(9): e1005871, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27636895

RESUMO

Human adenoviruses (HAdVs) are nonenveloped proteinaceous particles containing a linear double-stranded DNA genome. HAdVs cause a spectrum of pathologies in all populations regardless of health standards. Following repeat exposure to multiple HAdV types, we develop robust and long-lived humoral and cellular immune responses that provide life-long protection from de novo infections and persistent HAdV. How HAdVs, anti-HAdV antibodies and antigen presenting cells (APCs) interact to influence infection is still incompletely understood. In our study, we used physical, pharmacological, biochemical, fluorescence and electron microscopy, molecular and cell biology approaches to dissect the impact of immune-complexed HAdV (IC-HAdV) on human monocyte-derived dendritic cells (MoDCs). We show that IC-HAdV generate stabilized complexes of ~200 nm that are efficiently internalized by, and aggregate in, MoDCs. By comparing IC-HAdV, IC-empty capsid, IC-Ad2ts1 (a HAdV-C2 impaired in endosomal escape due to a mutation that impacts protease encapsidation) and IC-AdL40Q (a HAdV-C5 impaired in endosomal escape due to a mutation in protein VI), we demonstrate that protein VI-dependent endosomal escape is required for the HAdV genome to engage the DNA pattern recognition receptor AIM2 (absent in melanoma 2). AIM2 engagement induces pyroptotic MoDC death via ASC (apoptosis-associated speck protein containing a caspase activation/recruitment domain) aggregation, inflammasome formation, caspase 1 activation, and IL-1ß and gasdermin D (GSDMD) cleavage. Our study provides mechanistic insight into how humoral immunity initiates an innate immune response to HAdV-C5 in human professional APCs.


Assuntos
Infecções por Adenoviridae/imunologia , Adenovírus Humanos/imunologia , Complexo Antígeno-Anticorpo/imunologia , Proteínas de Ligação a DNA/imunologia , Piroptose/imunologia , Infecções por Adenoviridae/virologia , Adenovírus Humanos/genética , Caspase 1/metabolismo , Células Dendríticas/imunologia , Humanos , Imunidade Inata , Inflamassomos/imunologia , Interleucina-1beta/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Neoplasias/metabolismo , Proteínas de Ligação a Fosfato
16.
Nature ; 472(7344): 486-90, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21441909

RESUMO

CD4(+) T helper lymphocytes that express interleukin-17 (T(H)17 cells) have critical roles in mouse models of autoimmunity, and there is mounting evidence that they also influence inflammatory processes in humans. Genome-wide association studies in humans have linked genes involved in T(H)17 cell differentiation and function with susceptibility to Crohn's disease, rheumatoid arthritis and psoriasis. Thus, the pathway towards differentiation of T(H)17 cells and, perhaps, of related innate lymphoid cells with similar effector functions, is an attractive target for therapeutic applications. Mouse and human T(H)17 cells are distinguished by expression of the retinoic acid receptor-related orphan nuclear receptor RORγt, which is required for induction of IL-17 transcription and for the manifestation of T(H)17-dependent autoimmune disease in mice. By performing a chemical screen with an insect cell-based reporter system, we identified the cardiac glycoside digoxin as a specific inhibitor of RORγt transcriptional activity. Digoxin inhibited murine T(H)17 cell differentiation without affecting differentiation of other T cell lineages and was effective in delaying the onset and reducing the severity of autoimmune disease in mice. At high concentrations, digoxin is toxic for human cells, but non-toxic synthetic derivatives 20,22-dihydrodigoxin-21,23-diol and digoxin-21-salicylidene specifically inhibited induction of IL-17 in human CD4(+) T cells. Using these small-molecule compounds, we demonstrate that RORγt is important for the maintenance of IL-17 expression in mouse and human effector T cells. These data indicate that derivatives of digoxin can be used as chemical templates for the development of RORγt-targeted therapeutic agents that attenuate inflammatory lymphocyte function and autoimmune disease.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Digoxina/análogos & derivados , Digoxina/farmacologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/antagonistas & inibidores , Células Th17/citologia , Células Th17/efeitos dos fármacos , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Autoimunidade/efeitos dos fármacos , Autoimunidade/imunologia , Linhagem Celular , Digoxina/química , Digoxina/metabolismo , Digoxina/uso terapêutico , Drosophila/citologia , Humanos , Interleucina-17/biossíntese , Interleucina-17/imunologia , Camundongos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Células Th17/imunologia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
17.
Nature ; 467(7312): 214-7, 2010 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-20829794

RESUMO

Dendritic cells serve a key function in host defence, linking innate detection of microbes to activation of pathogen-specific adaptive immune responses. Whether there is cell-intrinsic recognition of human immunodeficiency virus (HIV) by host innate pattern-recognition receptors and subsequent coupling to antiviral T-cell responses is not yet known. Dendritic cells are largely resistant to infection with HIV-1, but facilitate infection of co-cultured T-helper cells through a process of trans-enhancement. Here we show that, when dendritic cell resistance to infection is circumvented, HIV-1 induces dendritic cell maturation, an antiviral type I interferon response and activation of T cells. This innate response is dependent on the interaction of newly synthesized HIV-1 capsid with cellular cyclophilin A (CYPA) and the subsequent activation of the transcription factor IRF3. Because the peptidylprolyl isomerase CYPA also interacts with HIV-1 capsid to promote infectivity, our results indicate that capsid conformation has evolved under opposing selective pressures for infectivity versus furtiveness. Thus, a cell-intrinsic sensor for HIV-1 exists in dendritic cells and mediates an antiviral immune response, but it is not typically engaged owing to the absence of dendritic cell infection. The virulence of HIV-1 may be related to evasion of this response, the manipulation of which may be necessary to generate an effective HIV-1 vaccine.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Infecções por HIV/imunologia , HIV-1/imunologia , Imunidade Inata , Proteínas do Capsídeo/imunologia , Linhagem Celular , Ciclofilina A/imunologia , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Infecções por HIV/virologia , HIV-1/fisiologia , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Ativação Linfocitária , Monócitos/citologia , Linfócitos T/imunologia
19.
J Cell Sci ; 126(Pt 24): 5553-65, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24105262

RESUMO

Exosomes are extracellular vesicles (EVs) secreted upon fusion of endosomal multivesicular bodies (MVBs) with the plasma membrane. The mechanisms involved in their biogenesis have not yet been fully identified although they could be used to modulate exosome formation and therefore are a promising tool in understanding exosome functions. We have performed an RNA interference screen targeting 23 components of the endosomal sorting complex required for transport (ESCRT) machinery and associated proteins in MHC class II (MHC II)-expressing HeLa-CIITA cells. Silencing of HRS, STAM1 or TSG101 reduced the secretion of EV-associated CD63 and MHC II but each gene altered differently the size and/or protein composition of secreted EVs, as quantified by immuno-electron microscopy. By contrast, depletion of VPS4B augmented this secretion while not altering the features of EVs. For several other ESCRT subunits, it was not possible to draw any conclusions about their involvement in exosome biogenesis from the screen. Interestingly, silencing of ALIX increased MHC II exosomal secretion, as a result of an overall increase in intracellular MHC II protein and mRNA levels. In human dendritic cells (DCs), ALIX depletion also increased MHC II in the cells, but not in the released CD63-positive EVs. Such differences could be attributed to a greater heterogeneity in size, and higher MHC II and lower CD63 levels in vesicles recovered from DCs as compared with HeLa-CIITA. The results reveal a role for selected ESCRT components and accessory proteins in exosome secretion and composition by HeLa-CIITA. They also highlight biogenetic differences in vesicles secreted by a tumour cell line and primary DCs.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Exossomos/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Dendríticas/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Técnicas de Silenciamento de Genes , Células HeLa , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Microscopia Imunoeletrônica , Corpos Multivesiculares/metabolismo , RNA Interferente Pequeno/genética , Tetraspanina 30/metabolismo
20.
Nat Commun ; 15(1): 4696, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38824133

RESUMO

Age-related microangiopathy, also known as small vessel disease (SVD), causes damage to the brain, retina, liver, and kidney. Based on the DNA damage theory of aging, we reasoned that genomic instability may underlie an SVD caused by dominant C-terminal variants in TREX1, the most abundant 3'-5' DNA exonuclease in mammals. C-terminal TREX1 variants cause an adult-onset SVD known as retinal vasculopathy with cerebral leukoencephalopathy (RVCL or RVCL-S). In RVCL, an aberrant, C-terminally truncated TREX1 mislocalizes to the nucleus due to deletion of its ER-anchoring domain. Since RVCL pathology mimics that of radiation injury, we reasoned that nuclear TREX1 would cause DNA damage. Here, we show that RVCL-associated TREX1 variants trigger DNA damage in humans, mice, and Drosophila, and that cells expressing RVCL mutant TREX1 are more vulnerable to DNA damage induced by chemotherapy and cytokines that up-regulate TREX1, leading to depletion of TREX1-high cells in RVCL mice. RVCL-associated TREX1 mutants inhibit homology-directed repair (HDR), causing DNA deletions and vulnerablility to PARP inhibitors. In women with RVCL, we observe early-onset breast cancer, similar to patients with BRCA1/2 variants. Our results provide a mechanistic basis linking aberrant TREX1 activity to the DNA damage theory of aging, premature senescence, and microvascular disease.


Assuntos
Dano ao DNA , Exodesoxirribonucleases , Fosfoproteínas , Animais , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Camundongos , Reparo de DNA por Recombinação , Fenótipo , Mutação , Drosophila/genética , Envelhecimento/genética , Envelhecimento/metabolismo , Feminino , Drosophila melanogaster/genética , Masculino , Doenças Retinianas , Doenças Vasculares , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA