Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(22): 4803-4817.e13, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37683634

RESUMEN

Patescibacteria, also known as the candidate phyla radiation (CPR), are a diverse group of bacteria that constitute a disproportionately large fraction of microbial dark matter. Its few cultivated members, belonging mostly to Saccharibacteria, grow as epibionts on host Actinobacteria. Due to a lack of suitable tools, the genetic basis of this lifestyle and other unique features of Patescibacteira remain unexplored. Here, we show that Saccharibacteria exhibit natural competence, and we exploit this property for their genetic manipulation. Imaging of fluorescent protein-labeled Saccharibacteria provides high spatiotemporal resolution of phenomena accompanying epibiotic growth, and a transposon-insertion sequencing (Tn-seq) genome-wide screen reveals the contribution of enigmatic Saccharibacterial genes to growth on their hosts. Finally, we leverage metagenomic data to provide cutting-edge protein structure-based bioinformatic resources that support the strain Southlakia epibionticum and its corresponding host, Actinomyces israelii, as a model system for unlocking the molecular underpinnings of the epibiotic lifestyle.


Asunto(s)
Bacterias , Bacterias/clasificación , Bacterias/genética , Bacterias/crecimiento & desarrollo , Metagenoma , Metagenómica , Filogenia , Actinobacteria/fisiología
2.
Trends Biochem Sci ; 48(6): 527-538, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37061423

RESUMEN

Protein-protein interactions (PPIs) drive biological processes, and disruption of PPIs can cause disease. With recent breakthroughs in structure prediction and a deluge of genomic sequence data, computational methods to predict PPIs and model spatial structures of protein complexes are now approaching the accuracy of experimental approaches for permanent interactions and show promise for elucidating transient interactions. As we describe here, the key to this success is rich evolutionary information deciphered from thousands of homologous sequences that coevolve in interacting partners. This covariation signal, revealed by sophisticated statistical and machine learning (ML) algorithms, predicts physiological interactions. Accurate artificial intelligence (AI)-based modeling of protein structures promises to provide accurate 3D models of PPIs at a proteome-wide scale.


Asunto(s)
Inteligencia Artificial , Mapeo de Interacción de Proteínas , Mapeo de Interacción de Proteínas/métodos , Algoritmos , Aprendizaje Automático , Proteoma , Biología Computacional/métodos
3.
Immunity ; 40(1): 40-50, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24412616

RESUMEN

Fibrosis in response to tissue damage or persistent inflammation is a pathological hallmark of many chronic degenerative diseases. By using a model of acute peritoneal inflammation, we have examined how repeated inflammatory activation promotes fibrotic tissue injury. In this context, fibrosis was strictly dependent on interleukin-6 (IL-6). Repeat inflammation induced IL-6-mediated T helper 1 (Th1) cell effector commitment and the emergence of STAT1 (signal transducer and activator of transcription-1) activity within the peritoneal membrane. Fibrosis was not observed in mice lacking interferon-γ (IFN-γ), STAT1, or RAG-1. Here, IFN-γ and STAT1 signaling disrupted the turnover of extracellular matrix by metalloproteases. Whereas IL-6-deficient mice resisted fibrosis, transfer of polarized Th1 cells or inhibition of MMP activity reversed this outcome. Thus, IL-6 causes compromised tissue repair by shifting acute inflammation into a more chronic profibrotic state through induction of Th1 cell responses as a consequence of recurrent inflammation.


Asunto(s)
Interleucina-6/metabolismo , Peritoneo/patología , Peritonitis/genética , Peritonitis/patología , Células TH1/inmunología , Enfermedad Aguda , Traslado Adoptivo , Animales , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Matriz Extracelular/inmunología , Retroalimentación Fisiológica , Fibrosis , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-6/genética , Interleucina-6/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Células TH1/trasplante
4.
Eur J Immunol ; 51(3): 742-745, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33125710

RESUMEN

Using a specific antibody, we found that expression of the viral restriction factor IFITM3 differs across cell types within the immune compartment with higher expression in myeloid rather than lymphoid cells. IFITM3 expression was increased following IFN stimulation, mostly type I, in immune cells, with the exception of T cells.


Asunto(s)
Antivirales/metabolismo , Interferón Tipo I/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Células A549 , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Linfocitos/metabolismo
5.
Mar Drugs ; 20(7)2022 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-35877711

RESUMEN

The mandelalides are complex macrolactone natural products with distinct macrocycle motifs and a bioactivity profile that is heavily influenced by compound glycosylation. Mandelalides A and B are direct inhibitors of mitochondrial ATP synthase (complex V) and therefore more toxic to mammalian cells with an oxidative metabolic phenotype. To provide further insight into the pharmacology of the mandelalides, we studied the AMP-activated protein kinase (AMPK) energy stress pathway and report that mandelalide A is an indirect activator of AMPK. Wild-type mouse embryonic fibroblasts (MEFs) and representative human non-small cell lung cancer (NSCLC) cells showed statistically significant increases in phospho-AMPK (Thr172) and phospho-ACC (Ser79) in response to mandelalide A. Mandelalide L, which also harbors an A-type macrocycle, induced similar increases in phospho-AMPK (Thr172) and phospho-ACC (Ser79) in U87-MG glioblastoma cells. In contrast, MEFs co-treated with an AMPK inhibitor (dorsomorphin), AMPKα-null MEFs, or NSCLC cells lacking liver kinase B1 (LKB1) lacked this activity. Mandelalide A was significantly more cytotoxic to AMPKα-null MEFs than wild-type cells, suggesting that AMPK activation serves as a protective response to mandelalide-induced depletion of cellular ATP. However, LKB1 status alone was not predictive of the antiproliferative effects of mandelalide A against NSCLC cells. When EGFR status was considered, erlotinib and mandelalide A showed strong cytotoxic synergy in combination against erlotinib-resistant 11-18 NSCLC cells but not against erlotinib-sensitive PC-9 cells. Finally, prolonged exposures rendered mandelalide A, a potent and efficacious cytotoxin, against a panel of human glioblastoma cell types regardless of the underlying metabolic phenotype of the cell. These results add biological relevance to the mandelalide series and provide the basis for their further pre-clinical evaluation as ATP synthase inhibitors and secondary activators of AMPK.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Glioblastoma , Neoplasias Pulmonares , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Clorhidrato de Erlotinib , Fibroblastos/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Macrólidos , Mamíferos/metabolismo , Ratones , Fosforilación
6.
Proteins ; 89(12): 1824-1833, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34324224

RESUMEN

For CASP14, we developed deep learning-based methods for predicting homo-oligomeric and hetero-oligomeric contacts and used them for oligomer modeling. To build structure models, we developed an oligomer structure generation method that utilizes predicted interchain contacts to guide iterative restrained minimization from random backbone structures. We supplemented this gradient-based fold-and-dock method with template-based and ab initio docking approaches using deep learning-based subunit predictions on 29 assembly targets. These methods produced oligomer models with summed Z-scores 5.5 units higher than the next best group, with the fold-and-dock method having the best relative performance. Over the eight targets for which this method was used, the best of the five submitted models had average oligomer TM-score of 0.71 (average oligomer TM-score of the next best group: 0.64), and explicit modeling of inter-subunit interactions improved modeling of six out of 40 individual domains (ΔGDT-TS > 2.0).


Asunto(s)
Modelos Moleculares , Conformación Proteica , Proteínas , Programas Informáticos , Biología Computacional , Bases de Datos de Proteínas , Aprendizaje Profundo , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas/química , Proteínas/metabolismo , Análisis de Secuencia de Proteína
7.
Proteins ; 89(12): 1722-1733, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34331359

RESUMEN

The trRosetta structure prediction method employs deep learning to generate predicted residue-residue distance and orientation distributions from which 3D models are built. We sought to improve the method by incorporating as inputs (in addition to sequence information) both language model embeddings and template information weighted by sequence similarity to the target. We also developed a refinement pipeline that recombines models generated by template-free and template utilizing versions of trRosetta guided by the DeepAccNet accuracy predictor. Both benchmark tests and CASP results show that the new pipeline is a considerable improvement over the original trRosetta, and it is faster and requires less computing resources, completing the entire modeling process in a median < 3 h in CASP14. Our human group improved results with this pipeline primarily by identifying additional homologous sequences for input into the network. We also used the DeepAccNet accuracy predictor to guide Rosetta high-resolution refinement for submissions in the regular and refinement categories; although performance was quite good on a CASP relative scale, the overall improvements were rather modest in part due to missing inter-domain or inter-chain contacts.


Asunto(s)
Biología Computacional/métodos , Aprendizaje Profundo , Estructura Terciaria de Proteína , Proteínas , Programas Informáticos , Humanos , Metagenoma/genética , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , Análisis de Secuencia de Proteína
8.
Immunology ; 164(2): 279-291, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34003499

RESUMEN

Cytomegalovirus (CMV) induction of large frequencies of highly functional memory T cells has attracted much interest in the utility of CMV-based vaccine vectors, with exciting preclinical data obtained in models of infectious diseases and cancer. However, pathogenesis of human CMV (HCMV) remains a concern. Attenuated CMV-based vectors, such as replication- or spread-deficient viruses, potentially offer an alternative to fully replicating vectors. However, it is not well understood how CMV attenuation impacts vector immunogenicity, particularly when administered via relevant routes of immunization such as the skin. Herein, we used the murine cytomegalovirus (MCMV) model to investigate the impact of vector attenuation on T-cell memory formation following subcutaneous administration. We found that the spread-deficient virus (ΔgL-MCMV) was impaired in its ability to induce memory CD8+ T cells reactive to some (M38, IE1) but not all (IE3) viral antigens. Impaired-memory T-cell development was associated with a preferential and pronounced loss of polyfunctional (IFN-γ+ TNF-α+ ) T cells and also reduced accumulation of TCF1+ T cells, and was not rescued by increasing the dose of replication-defective MCMV. Finally, whilst vector attenuation reduced dendritic cell (DC) recruitment to skin-draining lymph nodes, systematic depletion of multiple DC subsets during acute subcutaneous MCMV infection had a negligible impact on T-cell memory formation, implying that attenuated responses induced by replication-deficient vectors were likely not a consequence of impaired initial DC activation. Thus, overall, these data imply that the choice of antigen and/or cloning strategy of exogenous antigen in combination with the route of immunization may influence the ability of attenuated CMV vectors to induce robust functional T-cell memory.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Células Dendríticas/inmunología , Memoria Inmunológica/inmunología , Piel/inmunología , Replicación Viral/inmunología , Animales , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/virología , Infecciones por Citomegalovirus/virología , Células Dendríticas/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Piel/virología
9.
Thorax ; 76(12): 1246-1249, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34301738

RESUMEN

The burden of nosocomial SARS-CoV-2 infection remains poorly defined. We report on the outcomes of 2508 adults with molecularly-confirmed SARS-CoV-2 admitted across 18 major hospitals, representing over 60% of those hospitalised across Wales between 1 March and 1 July 2020. Inpatient mortality for nosocomial infection ranged from 38% to 42%, consistently higher than participants with community-acquired infection (31%-35%) across a range of case definitions. Those with hospital-acquired infection were older and frailer than those infected within the community. Nosocomial diagnosis occurred a median of 30 days following admission (IQR 21-63), suggesting a window for prophylactic or postexposure interventions, alongside enhanced infection control measures.


Asunto(s)
COVID-19 , Infección Hospitalaria , Adulto , Infección Hospitalaria/epidemiología , Hospitales , Humanos , Estudios Retrospectivos , SARS-CoV-2 , Gales/epidemiología
10.
J Virol ; 94(9)2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32075938

RESUMEN

Recognition of influenza A virus (IAV) by the innate immune system triggers pathways that restrict viral replication, activate innate immune cells, and regulate adaptive immunity. However, excessive innate immune activation can exaggerate disease. The pathways promoting excessive activation are incompletely understood, with limited experimental models to investigate the mechanisms driving influenza virus-induced inflammation in humans. Interferon regulatory factor 5 (IRF5) is a transcription factor that plays important roles in the induction of cytokines after viral sensing. In an in vivo model of IAV infection, IRF5 deficiency reduced IAV-driven immune pathology and associated inflammatory cytokine production, specifically reducing cytokine-producing myeloid cell populations in Irf5-/- mice but not impacting type 1 interferon (IFN) production or virus replication. Using cytometry by time of flight (CyTOF), we identified that human lung IRF5 expression was highest in cells of the myeloid lineage. To investigate the role of IRF5 in mediating human inflammatory responses by myeloid cells to IAV, we employed human-induced pluripotent stem cells (hIPSCs) with biallelic mutations in IRF5, demonstrating for the first time that induced pluripotent stem cell-derived dendritic cells (iPS-DCs) with biallelic mutations can be used to investigate the regulation of human virus-induced immune responses. Using this technology, we reveal that IRF5 deficiency in human DCs, or macrophages, corresponded with reduced virus-induced inflammatory cytokine production, with IRF5 acting downstream of Toll-like receptor 7 (TLR7) and, possibly, retinoic acid-inducible gene I (RIG-I) after viral sensing. Thus, IRF5 acts as a regulator of myeloid cell inflammatory cytokine production during IAV infection in mice and humans and drives immune-mediated viral pathogenesis independently of type 1 IFN and virus replication.IMPORTANCE The inflammatory response to influenza A virus (IAV) participates in infection control but contributes to disease severity. After viral detection, intracellular pathways are activated, initiating cytokine production, but these pathways are incompletely understood. We show that interferon regulatory factor 5 (IRF5) mediates IAV-induced inflammation and, in mice, drives pathology. This was independent of antiviral type 1 IFN and virus replication, implying that IRF5 could be specifically targeted to treat influenza virus-induced inflammation. We show for the first time that human iPSC technology can be exploited in genetic studies of virus-induced immune responses. Using this technology, we deleted IRF5 in human myeloid cells. These IRF5-deficient cells exhibited impaired influenza virus-induced cytokine production and revealed that IRF5 acts downstream of Toll-like receptor 7 and possibly retinoic acid-inducible gene I. Our data demonstrate the importance of IRF5 in influenza virus-induced inflammation, suggesting that genetic variation in the IRF5 gene may influence host susceptibility to viral diseases.


Asunto(s)
Células Madre Pluripotentes Inducidas/inmunología , Virus de la Influenza A/inmunología , Factores Reguladores del Interferón/metabolismo , Inmunidad Adaptativa/fisiología , Animales , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/fisiología , Virus de la Influenza A/metabolismo , Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Factores Reguladores del Interferón/inmunología , Interferón Tipo I/metabolismo , Pulmón/virología , Macrófagos/virología , Ratones , Infecciones por Orthomyxoviridae/virología , Replicación Viral/fisiología
11.
PLoS Pathog ; 15(6): e1007850, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31242262

RESUMEN

Invasive candidiasis, mainly caused by Candida albicans, is a serious healthcare problem with high mortality rates, particularly in immunocompromised patients. Innate immune cells express pathogen recognition receptors (PRRs) including C-type lectin-like receptors (CLRs) that bind C. albicans to initiate an immune response. Multiple CLRs including Dectin-1, Dectin-2 and Mincle have been proposed individually to contribute to the immune response to C. albicans. However how these receptors collaborate to clear a fungal infection is unknown. Herein, we used novel multi-CLR knockout (KO) mice to decipher the individual, collaborative and collective roles of Dectin-1, Dectin-2 and Mincle during systemic C. albicans infection. These studies revealed an unappreciated and profound role for CLR co-operation in anti-fungal immunity. The protective effect of multiple CLRs was markedly greater than any single receptor, and was mediated through inflammatory monocytes via recognition and phagocytosis of C. albicans, and production of C. albicans-induced cytokines and chemokines. These CLRs were dispensable for mediating similar responses from neutrophils, likely due to lower expression of these CLRs on neutrophils compared to inflammatory monocytes. Concurrent deletion of Dectin-1 and Dectin-2, or all three CLRs, resulted in dramatically increased susceptibility to systemic C. albicans infection compared to mice lacking a single CLR. Multi-CLR KO mice were unable to control fungal growth due to an inadequate early inflammatory monocyte-mediated response. In response to excessive fungal growth, the multi-CLR KO mice mounted a hyper-inflammatory response, likely leading to multiple organ failure. Thus, these data reveal a critical role for CLR co-operation in the effective control of C. albicans and maintenance of organ function during infection.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Lectinas Tipo C/inmunología , Proteínas de la Membrana/inmunología , Monocitos/inmunología , Animales , Candidiasis/genética , Quimiocinas/genética , Quimiocinas/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Lectinas Tipo C/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Monocitos/patología , Neutrófilos/patología
12.
J Immunol ; 202(3): 943-955, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30635396

RESUMEN

Candidate vaccines designed to generate T cell-based immunity are typically vectored by nonpersistent viruses, which largely fail to elicit durable effector memory T cell responses. This limitation can be overcome using recombinant strains of CMV. Proof-of-principle studies have demonstrated the potential benefits of this approach, most notably in the SIV model, but safety concerns require the development of nonreplicating alternatives with comparable immunogenicity. In this study, we show that IL-33 promotes the accumulation and recall kinetics of circulating and tissue-resident memory T cells in mice infected with murine CMV. Using a replication-deficient murine CMV vector, we further show that exogenous IL-33 boosts vaccine-induced memory T cell responses, which protect against subsequent heterologous viral challenge. These data suggest that IL-33 could serve as a useful adjuvant to improve the efficacy of vaccines based on attenuated derivatives of CMV.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra Citomegalovirus/inmunología , Memoria Inmunológica , Interleucina-33/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Citomegalovirus , Interleucina-33/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Muromegalovirus , Vacunas Atenuadas/inmunología
13.
Nucleic Acids Res ; 45(20): 11673-11683, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-28981850

RESUMEN

The HIRA histone chaperone complex deposits histone H3.3 into nucleosomes in a DNA replication- and sequence-independent manner. As herpesvirus genomes enter the nucleus as naked DNA, we asked whether the HIRA chaperone complex affects herpesvirus infection. After infection of primary cells with HSV or CMV, or transient transfection with naked plasmid DNA, HIRA re-localizes to PML bodies, sites of cellular anti-viral activity. HIRA co-localizes with viral genomes, binds to incoming viral and plasmid DNAs and deposits histone H3.3 onto these. Anti-viral interferons (IFN) specifically induce HIRA/PML co-localization at PML nuclear bodies and HIRA recruitment to IFN target genes, although HIRA is not required for IFN-inducible expression of these genes. HIRA is, however, required for suppression of viral gene expression, virus replication and lytic infection and restricts murine CMV replication in vivo. We propose that the HIRA chaperone complex represses incoming naked viral DNAs through chromatinization as part of intrinsic cellular immunity.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , ADN Viral/metabolismo , Herpesvirus Humano 1/metabolismo , Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Línea Celular , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Cromatina/virología , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , ADN Viral/genética , Células HEK293 , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Chaperonas de Histonas/genética , Chaperonas de Histonas/inmunología , Humanos , Cuerpos de Inclusión/inmunología , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/virología , Ratones Endogámicos C57BL , Muromegalovirus/genética , Muromegalovirus/fisiología , Proteína de la Leucemia Promielocítica/metabolismo , Unión Proteica , Factores de Transcripción/genética , Factores de Transcripción/inmunología
14.
Immunology ; 153(1): 1-9, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28869761

RESUMEN

Since the development of vaccinia virus as a vaccine vector in 1984, the utility of numerous viruses in vaccination strategies has been explored. In recent years, key improvements to existing vectors such as those based on adenovirus have led to significant improvements in immunogenicity and efficacy. Furthermore, exciting new vectors that exploit viruses such as cytomegalovirus (CMV) and vesicular stomatitis virus (VSV) have emerged. Herein, we summarize these recent developments in viral vector technologies, focusing on novel vectors based on CMV, VSV, measles and modified adenovirus. We discuss the potential utility of these exciting approaches in eliciting protection against infectious diseases.


Asunto(s)
Control de Enfermedades Transmisibles , Enfermedades Transmisibles/inmunología , Vectores Genéticos/genética , Vacunas Virales/genética , Vacunas Virales/inmunología , Virus/genética , Inmunidad Adaptativa , Animales , Control de Enfermedades Transmisibles/métodos , Vectores Genéticos/inmunología , Humanos , Vacunación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Virus/clasificación , Virus/inmunología
15.
Genome Res ; 25(2): 226-34, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25367293

RESUMEN

V(D)J genomic recombination joins single gene segments to encode an extensive repertoire of antigen receptor specificities in T and B lymphocytes. This process initiates with double-stranded breaks adjacent to conserved recombination signal sequences that contain either 12- or 23-nucleotide spacer regions. Only recombination between signal sequences with unequal spacers results in productive coding genes, a phenomenon known as the "12/23 rule." Here we present two novel genomic tools that allow the capture and analysis of immune locus rearrangements from whole thymic and splenic tissues using second-generation sequencing. Further, we provide strong evidence that the 12/23 rule of genomic recombination is frequently violated under physiological conditions, resulting in unanticipated hybrid recombinations in ∼10% of Tcra excision circles. Hence, we demonstrate that strict adherence to the 12/23 rule is intrinsic neither to recombination signal sequences nor to the catalytic process of recombination and propose that nonclassical excision circles are liberated during the formation of antigen receptor diversity.


Asunto(s)
Genómica , Linfocitos/metabolismo , Recombinación V(D)J , Animales , Diferenciación Celular/genética , Reordenamiento Génico , Genes RAG-1 , Humanos , Linfocitos/citología , Ratones , Ratones Noqueados , Modelos Biológicos , Análisis de Secuencia de ADN
16.
PLoS Pathog ; 12(12): e1006050, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27926930

RESUMEN

CD4+ T cells support host defence against herpesviruses and other viral pathogens. We identified that CD4+ T cells from systemic and mucosal tissues of hosts infected with the ß-herpesviridae human cytomegalovirus (HCMV) or murine cytomegalovirus (MCMV) express the regulatory cytokine interleukin (IL)-10. IL-10+CD4+ T cells co-expressed TH1-associated transcription factors and chemokine receptors. Mice lacking T cell-derived IL-10 elicited enhanced antiviral T cell responses and restricted MCMV persistence in salivary glands and secretion in saliva. Thus, IL-10+CD4+ T cells suppress antiviral immune responses against CMV. Expansion of this T-cell population in the periphery was promoted by IL-27 whereas mucosal IL-10+ T cell responses were ICOS-dependent. Infected Il27rα-deficient mice with reduced peripheral IL-10+CD4+ T cell accumulation displayed robust T cell responses and restricted MCMV persistence and shedding. Temporal inhibition experiments revealed that IL-27R signaling during initial infection was required for the suppression of T cell immunity and control of virus shedding during MCMV persistence. IL-27 production was promoted by type-I IFN, suggesting that ß-herpesviridae exploit the immune-regulatory properties of this antiviral pathway to establish chronicity. Further, our data reveal that cytokine signaling events during initial infection profoundly influence virus chronicity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Interferón Tipo I/inmunología , Interleucina-27/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Interleucina-10/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
J Immunol ; 197(1): 208-21, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27259855

RESUMEN

Candida spp. elicit cytokine production downstream of various pathogen recognition receptors, including C-type lectin-like receptors, TLRs, and nucleotide oligomerization domain (NOD)-like receptors. IL-12 family members IL-12p70 and IL-23 are important for host immunity against Candida spp. In this article, we show that IL-27, another IL-12 family member, is produced by myeloid cells in response to selected Candida spp. We demonstrate a novel mechanism for Candida parapsilosis-mediated induction of IL-27 in a TLR7-, MyD88-, and NOD2-dependent manner. Our data revealed that IFN-ß is induced by C. parapsilosis, which in turn signals through the IFN-α/ß receptor and STAT1/2 to induce IL-27. Moreover, IL-27R (WSX-1)-deficient mice systemically infected with C. parapsilosis displayed enhanced pathogen clearance compared with wild-type mice. This was associated with increased levels of proinflammatory cytokines in the serum and increased IFN-γ and IL-17 responses in the spleens of IL-27R-deficient mice. Thus, our data define a novel link between C. parapsilosis, TLR7, NOD2, IFN-ß, and IL-27, and we have identified an important role for IL-27 in the immune response against C. parapsilosis Overall, these findings demonstrate an important mechanism for the suppression of protective immune responses during infection with C. parapsilosis, which has potential relevance for infections with other fungal pathogens.


Asunto(s)
Candida/fisiología , Candidiasis/inmunología , Interleucina-27/metabolismo , Células Mieloides/inmunología , Receptor Toll-Like 7/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Evasión Inmune , Mediadores de Inflamación/metabolismo , Interferón beta/metabolismo , Interleucina-27/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , Receptores de Citocinas/genética , Receptores de Interleucina , Transducción de Señal
18.
Mar Drugs ; 16(3)2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29494533

RESUMEN

Our understanding of autophagy and lysosomal function has been greatly enhanced by the discovery of natural product structures that can serve as chemical probes to reveal new patterns of signal transduction in cells. Coibamide A is a cytotoxic marine natural product that induces mTOR-independent autophagy as an adaptive stress response that precedes cell death. Autophagy-related (ATG) protein 5 (ATG5) is required for coibamide-induced autophagy but not required for coibamide-induced apoptosis. Using wild-type and autophagy-deficient mouse embryonic fibroblasts (MEFs) we demonstrate that coibamide-induced toxicity is delayed in ATG5-/- cells relative to ATG5+/+ cells. Time-dependent changes in annexin V staining, membrane integrity, metabolic capacity and caspase activation indicated that MEFs with a functional autophagy pathway are more sensitive to coibamide A. This pattern could be distinguished from autophagy modulators that induce acute ER stress (thapsigargin, tunicamycin), ATP depletion (oligomycin A) or mTORC1 inhibition (rapamycin), but was shared with the Sec61 inhibitor apratoxin A. Coibamide- or apratoxin-induced cell stress was further distinguished from the action of thapsigargin by a pattern of early LC3-II accumulation in the absence of CHOP or BiP expression. Time-dependent changes in ATG5-ATG12, PARP1 and caspase-3 expression patterns were consistent with the conversion of ATG5 to a pro-death signal in response to both compounds.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/metabolismo , Autofagia/efectos de los fármacos , Depsipéptidos/toxicidad , Animales , Proteína 5 Relacionada con la Autofagia/genética , Línea Celular , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Fibroblastos , Técnicas de Inactivación de Genes , Toxinas Marinas/toxicidad , Ratones , Transducción de Señal/efectos de los fármacos , Tapsigargina/toxicidad
19.
Eur J Immunol ; 46(9): 2222-32, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27378515

RESUMEN

The inflammatory activation and recruitment of defined myeloid populations is essential for controlling the bridge between innate and adaptive immunity and shaping the immune response to microbial challenge. However, these cells exhibit significant functional heterogeneity and the inflammatory signals that differentially influence their effector characteristics are poorly characterized. In this study, we defined the phenotype of discrete subsets of effective antigen-presenting cells (APCs) in the peritoneal cavity during peritonitis. When the functional properties of these cells were compared to inflammatory monocyte-derived macrophages we noted differential responses to the immune-modulatory cytokine IL-10. In contrast to the suppressive actions of IL-10 on inflammatory macrophages, the recruitment of APCs was relatively refractory and we found no evidence for selective inhibition of APC differentiation. This differential response of myeloid cell subsets to IL-10 may thus have limited impact on development of potentially tissue-damaging adaptive immune responses, while restricting the magnitude of the inflammatory response. These findings may have clinical relevance in the context of peritoneal dialysis patients, where recurrent infections are associated with immune-mediated membrane dysfunction, treatment failure, and increased morbidity.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-10/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Animales , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/patología , Biomarcadores , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Inmunomodulación , Inmunofenotipificación , Inflamación/patología , Interleucina-10/genética , Macrófagos/patología , Ratones , Ratones Noqueados , Peritonitis/inmunología , Peritonitis/metabolismo , Peritonitis/patología , Fenotipo , Receptores CCR2/metabolismo
20.
PLoS Pathog ; 11(2): e1004641, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25654642

RESUMEN

CD200 receptor (CD200R) negatively regulates peripheral and mucosal innate immune responses. Viruses, including herpesviruses, have acquired functional CD200 orthologs, implying that viral exploitation of this pathway is evolutionary advantageous. However, the role that CD200R signaling plays during herpesvirus infection in vivo requires clarification. Utilizing the murine cytomegalovirus (MCMV) model, we demonstrate that CD200R facilitates virus persistence within mucosal tissue. Specifically, MCMV infection of CD200R-deficient mice (CD200R(-/-)) elicited heightened mucosal virus-specific CD4 T cell responses that restricted virus persistence in the salivary glands. CD200R did not directly inhibit lymphocyte effector function. Instead, CD200R(-/-) mice exhibited enhanced APC accumulation that in the mucosa was a consequence of elevated cellular proliferation. Although MCMV does not encode an obvious CD200 homolog, productive replication in macrophages induced expression of cellular CD200. CD200 from hematopoietic and non-hematopoietic cells contributed independently to suppression of antiviral control in vivo. These results highlight the CD200-CD200R pathway as an important regulator of antiviral immunity during cytomegalovirus infection that is exploited by MCMV to establish chronicity within mucosal tissue.


Asunto(s)
Antígenos CD/inmunología , Infecciones por Citomegalovirus/inmunología , Macrófagos/inmunología , Membrana Mucosa/inmunología , Membrana Mucosa/virología , Animales , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Macrófagos/metabolismo , Macrófagos/virología , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA