Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Nat Commun ; 15(1): 4696, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38824133

RESUMEN

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.


Asunto(s)
Daño del ADN , Exodesoxirribonucleasas , Fosfoproteínas , Animales , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ratones , Reparación del ADN por Recombinación , Fenotipo , Mutación , Drosophila/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Femenino , Drosophila melanogaster/genética , Masculino , Enfermedades de la Retina , Enfermedades Vasculares , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias
2.
Nat Aging ; 3(10): 1251-1268, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37723209

RESUMEN

Aging is characterized by gradual immune dysfunction and increased disease risk. Genomic instability is considered central to the aging process, but the underlying mechanisms of DNA damage are insufficiently defined. Cells in confined environments experience forces applied to their nucleus, leading to transient nuclear envelope rupture (NER) and DNA damage. Here, we show that Lamin A/C protects lung alveolar macrophages (AMs) from NER and hallmarks of aging. AMs move within constricted spaces in the lung. Immune-specific ablation of lamin A/C results in selective depletion of AMs and heightened susceptibility to influenza virus-induced pathogenesis and lung cancer growth. Lamin A/C-deficient AMs that persist display constitutive NER marks, DNA damage and p53-dependent senescence. AMs from aged wild-type and from lamin A/C-deficient mice share a lysosomal signature comprising CD63. CD63 is required to limit damaged DNA in macrophages. We propose that NER-induced genomic instability represents a mechanism of aging in AMs.


Asunto(s)
Lamina Tipo A , Macrófagos Alveolares , Animales , Ratones , Lamina Tipo A/genética , Membrana Nuclear , Pulmón , Envejecimiento/genética , Inestabilidad Genómica
3.
Nature ; 617(7960): 386-394, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37100912

RESUMEN

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.


Asunto(s)
Plasticidad de la Célula , Cobre , Inflamación , Transducción de Señal , Animales , Ratones , Cobre/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , NAD/metabolismo , Transducción de Señal/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Peróxido de Hidrógeno/metabolismo , Epigénesis Genética/efectos de los fármacos , Metformina/análogos & derivados , Oxidación-Reducción , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/genética , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/genética
4.
J Exp Med ; 220(5)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36820829

RESUMEN

In innate immune cells, intracellular sensors such as cGAS-STING stimulate type I/III interferon (IFN) expression, which promotes antiviral defense and immune activation. However, how IFN-I/III expression is controlled in adaptive cells is poorly understood. Here, we identify a transcriptional rheostat orchestrated by RELA that confers human T cells with innate-like abilities to produce IFN-I/III. Despite intact cGAS-STING signaling, IFN-I/III responses are stunted in CD4+ T cells compared with dendritic cells or macrophages. We find that lysine residues in RELA tune the IFN-I/III response at baseline and in response to STING stimulation in CD4+ T cells. This response requires positive feedback driven by cGAS and IRF7 expression. By combining RELA with IRF3 and DNA demethylation, IFN-I/III production in CD4+ T cells reaches levels observed in dendritic cells. IFN-I/III production provides self-protection of CD4+ T cells against HIV infection and enhances the elimination of tumor cells by CAR T cells. Therefore, innate-like functions can be tuned and leveraged in human T cells.


Asunto(s)
Infecciones por VIH , Interferón Tipo I , Humanos , Inmunidad Innata/genética , Nucleotidiltransferasas/metabolismo , Interferón Tipo I/metabolismo , Linfocitos T/metabolismo , Factor de Transcripción ReIA
5.
Sci Immunol ; 8(79): eabn6612, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36638189

RESUMEN

T cells that recognize tumor antigens are crucial for mounting antitumor immune responses. Induction of antitumor T cells in immunogenic tumors depends on STING, the intracellular innate immune receptor for cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) and related cyclic dinucleotides (CDNs). However, the optimal way to leverage STING activation in nonimmunogenic tumors is still unclear. Here, we show that cGAMP delivery by intratumoral injection of virus-like particles (cGAMP-VLP) led to differentiation of circulating tumor-specific T cells, decreased tumor regulatory T cells (Tregs), and antitumoral responses that synergized with PD1 blockade. By contrast, intratumoral injection of the synthetic CDN ADU-S100 led to tumor necrosis and systemic T cell activation but simultaneously depleted immune cells from injected tumors and induced minimal priming of circulating tumor-specific T cells. The antitumor effects of cGAMP-VLP required type 1 conventional dendritic cells (cDC1), whereas ADU-S100 eliminated cDC1 from injected tumors. cGAMP-VLP preferentially targeted STING in dendritic cells at a 1000-fold smaller dose than ADU-S100. Subcutaneous administration of cGAMP-VLP showed synergy when combined with PD1 blockade or a tumor Treg-depleting antibody to elicit systemic tumor-specific T cells and antitumor activity, leading to complete and durable tumor eradication in the case of tumor Treg depletion. These findings show that cell targeting of STING stimulation shapes the antitumor T cell response and identify a therapeutic strategy to enhance T cell-targeted immunotherapy.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Inmunidad , Células Dendríticas
6.
J Exp Med ; 220(1)2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36346347

RESUMEN

Defects in nucleic acid metabolizing enzymes can lead to spontaneous but selective activation of either cGAS/STING or RIG-like receptor (RLR) signaling, causing type I interferon-driven inflammatory diseases. In these pathophysiological conditions, activation of the DNA sensor cGAS and IFN production are linked to spontaneous DNA damage. Physiological, or tonic, IFN signaling on the other hand is essential to functionally prime nucleic acid sensing pathways. Here, we show that low-level chronic DNA damage in mice lacking the Aicardi-Goutières syndrome gene SAMHD1 reduced tumor-free survival when crossed to a p53-deficient, but not to a DNA mismatch repair-deficient background. Increased DNA damage did not result in higher levels of type I interferon. Instead, we found that the chronic interferon response in SAMHD1-deficient mice was driven by the MDA5/MAVS pathway but required functional priming through the cGAS/STING pathway. Our work positions cGAS/STING upstream of tonic IFN signaling in Samhd1-deficient mice and highlights an important role of the pathway in physiological and pathophysiological innate immune priming.


Asunto(s)
Interferón Tipo I , Ácidos Nucleicos , Ratones , Animales , Proteína 1 que Contiene Dominios SAM y HD/genética , Inmunidad Innata/genética , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Interferón Tipo I/metabolismo
7.
Proc Natl Acad Sci U S A ; 119(17): e2107394119, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35439048

RESUMEN

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.


Asunto(s)
Vesículas Extracelulares , Neoplasias de la Mama Triple Negativas , Vesículas Extracelulares/fisiología , Humanos , Macrófagos , Neoplasias de la Mama Triple Negativas/patología
8.
Cell ; 184(20): 5230-5246.e22, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34551315

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Daño del ADN , Exodesoxirribonucleasas/metabolismo , Membrana Nuclear/metabolismo , Fosfoproteínas/metabolismo , Animales , Línea Celular , Senescencia Celular , Colágeno/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Ratones , Invasividad Neoplásica , Membrana Nuclear/ultraestructura , Proteolisis , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Sci Signal ; 14(697)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34429383

RESUMEN

Modified vaccinia Ankara (MVA) is a live, attenuated human smallpox vaccine and a vector for the development of new vaccines against infectious diseases and cancer. Efficient activation of the immune system by MVA partially relies on its encounter with dendritic cells (DCs). MVA infection of DCs leads to multiple outcomes, including cytokine production, activation of costimulatory molecules for T cell stimulation, and cell death. Here, we examined how these diverse responses are orchestrated in human DCs. Single-cell analyses revealed that the response to MVA infection in DCs was limited to early viral gene expression. In response to the early events in the viral cycle, we found that DCs grouped into three distinct clusters. A cluster of infected cells sensed the MVA genome by the intracellular innate immunity pathway mediated by cGAS, STING, TBK1, and IRF3 and subsequently produced inflammatory cytokines. In response to these cytokines, a cluster of noninfected bystander cells increased costimulatory molecule expression. A separate cluster of infected cells underwent caspase-dependent apoptosis. Induction of apoptosis persisted after inhibition of innate immunity pathway mediators independently of previously described IRF-dependent or replication-dependent pathways and was a response to early MVA gene expression. Together, our study identified multiple mechanisms that underlie the interactions of MVA with human DCs.


Asunto(s)
Vaccinia , Vacunas Virales , Células Dendríticas , Humanos , Análisis de la Célula Individual , Vacunas de ADN
10.
EMBO J ; 39(20): e105505, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32945564

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Proteína A Centromérica/metabolismo , Proteína B del Centrómero/metabolismo , Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica , Nucleosomas/genética , Linfocitos T CD4-Positivos/citología , Sistemas CRISPR-Cas , Ciclo Celular , Línea Celular Tumoral , Centrómero/genética , Segregación Cromosómica/genética , Biología Computacional , Epigénesis Genética , Marcación de Gen , Humanos , Hibridación Fluorescente in Situ , Nucleosomas/metabolismo , ARN Interferente Pequeño
11.
Innate Immun ; 26(6): 459-472, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32248720

RESUMEN

A subset of TLRs is specialised in the detection of incoming pathogens by sampling endosomes for nucleic acid contents. Among them, TLR3 senses the abnormal presence of double-stranded RNA in the endosomes and initiates a potent innate immune response via activation of NF-κB and IRF3. Nevertheless, mechanisms governing TLR3 regulation remain poorly defined. To identify new molecular players involved in the TLR3 pathway, we performed a genome-wide screen using CRISPR/Cas9 technology. We generated TLR3+ reporter cells carrying a NF-κB-responsive promoter that controls GFP expression. Cells were next transduced with a single-guide RNA (sgRNA) library, subjected to sequential rounds of stimulation with poly(I:C) and sorting of the GFP-negative cells. Enrichments in sgRNA estimated by deep sequencing identified genes required for TLR3-induced activation of NF-κB. Among the hits, five genes known to be critically involved in the TLR3 pathway, including TLR3 itself and the chaperone UNC93B1, were identified by the screen, thus validating our strategy. We further studied the top 40 hits and focused on the transcription factor aryl hydrocarbon receptor (AhR). Depletion of AhR had a dual effect on the TLR3 response, abrogating IL-8 production and enhancing IP-10 release. Moreover, in primary human macrophages exposed to poly(I:C), AhR activation enhanced IL-8 and diminished IP-10 release. Overall, these results reveal AhR plays a role in the TLR3 cellular innate immune response.


Asunto(s)
Macrófagos/inmunología , Receptores de Hidrocarburo de Aril/genética , Receptor Toll-Like 3/metabolismo , Células Cultivadas , Quimiocina CXCL10/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interleucina-8/metabolismo , Proteínas de Transporte de Membrana/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Poli I-C/inmunología , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Receptor Toll-Like 3/genética
12.
J Exp Med ; 216(5): 1199-1213, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-30936263

RESUMEN

Cellular innate immune sensors of DNA are essential for host defense against invading pathogens. However, the presence of self-DNA inside cells poses a risk of triggering unchecked immune responses. The mechanisms limiting induction of inflammation by self-DNA are poorly understood. BLM RecQ-like helicase is essential for genome integrity and is deficient in Bloom syndrome (BS), a rare genetic disease characterized by genome instability, accumulation of micronuclei, susceptibility to cancer, and immunodeficiency. Here, we show that BLM-deficient fibroblasts show constitutive up-regulation of inflammatory interferon-stimulated gene (ISG) expression, which is mediated by the cGAS-STING-IRF3 cytosolic DNA-sensing pathway. Increased DNA damage or down-regulation of the cytoplasmic exonuclease TREX1 enhances ISG expression in BLM-deficient fibroblasts. cGAS-containing cytoplasmic micronuclei are increased in BS cells. Finally, BS patients demonstrate elevated ISG expression in peripheral blood. These results reveal that BLM limits ISG induction, thus connecting DNA damage to cellular innate immune response, which may contribute to human pathogenesis.


Asunto(s)
Inmunidad Innata/inmunología , Nucleotidiltransferasas/metabolismo , RecQ Helicasas/inmunología , 2',5'-Oligoadenilato Sintetasa/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Síndrome de Bloom/patología , Niño , Citosol/metabolismo , Daño del ADN/inmunología , Exodesoxirribonucleasas/metabolismo , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Factor 3 Regulador del Interferón/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/genética , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/genética , RecQ Helicasas/genética , Transcriptoma , Transducción Genética , Proteínas Supresoras de Tumor/genética
13.
Cell Rep ; 26(9): 2377-2393.e13, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30811988

RESUMEN

Cytosolic DNA activates cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS), an innate immune sensor pivotal in anti-microbial defense, senescence, auto-immunity, and cancer. cGAS is considered to be a sequence-independent DNA sensor with limited access to nuclear DNA because of compartmentalization. However, the nuclear envelope is a dynamic barrier, and cGAS is present in the nucleus. Here, we identify determinants of nuclear cGAS localization and activation. We show that nuclear-localized cGAS synthesizes cGAMP and induces innate immune activation of dendritic cells, although cGAMP levels are 200-fold lower than following transfection with exogenous DNA. Using cGAS ChIP-seq and a GFP-cGAS knockin mouse, we find nuclear cGAS enrichment on centromeric satellite DNA, confirmed by imaging, and to a lesser extent on LINE elements. The non-enzymatic N-terminal domain of cGAS determines nucleo-cytoplasmic localization, enrichment on centromeres, and activation of nuclear-localized cGAS. These results reveal a preferential functional association of nuclear cGAS with centromeres.


Asunto(s)
Centrómero/enzimología , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Inmunidad Innata/genética , Nucleotidiltransferasas/metabolismo , Adulto , Animales , Línea Celular , Núcleo Celular/enzimología , ADN , ADN Satélite , Femenino , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Nucleotidiltransferasas/química , Dominios Proteicos
14.
Cell ; 175(2): 488-501.e22, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30270045

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/inmunología , Proteínas Asociadas a Matriz Nuclear/inmunología , Proteínas Asociadas a Matriz Nuclear/fisiología , Factores de Transcripción de Octámeros/inmunología , Factores de Transcripción de Octámeros/fisiología , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/fisiología , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/fisiología , Núcleo Celular/metabolismo , ADN Viral/genética , ADN Viral/inmunología , Proteínas de Unión al ADN , Células Dendríticas/inmunología , Infecciones por VIH/inmunología , VIH-1/genética , VIH-1/inmunología , VIH-2/genética , VIH-2/inmunología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata/inmunología , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/fisiología , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/inmunología
15.
Hepatology ; 68(5): 1695-1709, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29679386

RESUMEN

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.


Asunto(s)
Virus de la Hepatitis B/patogenicidad , Hepatitis B/fisiopatología , Hepatocitos/virología , Evasión Inmune/fisiología , Nucleótidos Cíclicos/metabolismo , Animales , Western Blotting , Técnicas de Cultivo de Célula , ADN Viral/inmunología , Perfilación de la Expresión Génica/métodos , Hepatitis B/inmunología , Hepatocitos/metabolismo , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune/inmunología , Hibridación Fluorescente in Situ/métodos , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
Cytokine Growth Factor Rev ; 40: 59-67, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29526438

RESUMEN

The nuclear envelope is a physical barrier that isolates the cellular DNA from the rest of the cell, thereby limiting pathogen invasion. The Human Immunodeficiency Virus (HIV) has a remarkable ability to enter the nucleus of non-dividing target cells such as lymphocytes, macrophages and dendritic cells. While this step is critical for replication of the virus, it remains one of the less understood aspects of HIV infection. Here, we review the viral and host factors that favor or inhibit HIV entry into the nucleus, including the viral capsid, integrase, the central viral DNA flap, and the host proteins CPSF6, TNPO3, Nucleoporins, SUN1, SUN2, Cyclophilin A and MX2. We review recent perspectives on the mechanism of action of these factors, and formulate fundamental questions that remain. Overall, these findings deepen our understanding of HIV nuclear import and strengthen the favorable position of nuclear HIV entry for antiviral targeting.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Infecciones por VIH/patología , VIH-1/fisiología , Membrana Nuclear/fisiología , Integración Viral/fisiología , Replicación Viral/fisiología , Células Dendríticas/virología , Interacciones Huésped-Patógeno/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfocitos/virología , Macrófagos/virología , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Nucleares/metabolismo
17.
Annu Rev Immunol ; 36: 193-220, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29328787

RESUMEN

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.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Retroviridae/inmunología , Inmunidad Adaptativa , Animales , Humanos , Evasión Inmune , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunidad Innata , Infecciones por Retroviridae/metabolismo
18.
Infect Genet Evol ; 55: 142-150, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28919545

RESUMEN

Immune evasion is critical for pathogens to maintain their presence within hosts, giving rise to chronic infections. Here, we examine the immune evasion strategies employed by three pathogens with high medical burden, namely, tuberculosis, HIV and HBV. Establishment of chronic infection by these pathogens is a multi-step process that involves an interplay between restriction factor, innate immunity and adaptive immunity. Engagement of these host defences is intimately linked with specific steps within the pathogen replication cycles. Critical host factors are increasingly recognized to regulate immune evasion and susceptibility to disease. Fuelled by innovative technology development, the understanding of these mechanisms provides critical knowledge for rational design of vaccines and therapeutic immune strategies.


Asunto(s)
Infecciones por VIH/etiología , VIH/inmunología , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/etiología , Interacciones Huésped-Patógeno/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/etiología , Humanos , Evasión Inmune
19.
PLoS Pathog ; 12(9): e1005871, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27636895

RESUMEN

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.


Asunto(s)
Infecciones por Adenoviridae/inmunología , Adenovirus Humanos/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Proteínas de Unión al ADN/inmunología , Piroptosis/inmunología , Infecciones por Adenoviridae/virología , Adenovirus Humanos/genética , Caspasa 1/metabolismo , Células Dendríticas/inmunología , Humanos , Inmunidad Innata , Inflamasomas/inmunología , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Neoplasias/metabolismo , Proteínas de Unión a Fosfato
20.
Science ; 349(6253): 1232-6, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26229115

RESUMEN

Infected cells detect viruses through a variety of receptors that initiate cell-intrinsic innate defense responses. Cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase (cGAS) is a cytosolic sensor for many DNA viruses and HIV-1. In response to cytosolic viral DNA, cGAS synthesizes the second messenger 2'3'-cyclic GMP-AMP (cGAMP), which activates antiviral signaling pathways. We show that in cells producing virus, cGAS-synthesized cGAMP can be packaged in viral particles and extracellular vesicles. Viral particles efficiently delivered cGAMP to target cells. cGAMP transfer by viral particles to dendritic cells activated innate immunity and antiviral defenses. Finally, we show that cell-free murine cytomegalovirus and Modified Vaccinia Ankara virus contained cGAMP. Thus, transfer of cGAMP by viruses may represent a defense mechanism to propagate immune responses to uninfected target cells.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Herpesviridae/inmunología , Inmunidad Innata/inmunología , Muromegalovirus/metabolismo , Nucleótidos Cíclicos/metabolismo , Sistemas de Mensajero Secundario , Virus Vaccinia/metabolismo , Vaccinia/inmunología , Virión/metabolismo , Animales , Chlorocebus aethiops , Citosol/inmunología , Citosol/metabolismo , Citosol/virología , Células Dendríticas/virología , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Infecciones por VIH/inmunología , VIH-1/genética , VIH-1/metabolismo , Células HeLa , Humanos , Inmunidad Innata/genética , Ratones , Ratones Endogámicos C57BL , Muromegalovirus/genética , Virus Vaccinia/genética , Células Vero , Virión/genética , Ensamble de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA