Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(51): e2300474120, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38100417

RESUMEN

Seasonal influenza results in 3 to 5 million cases of severe disease and 250,000 to 500,000 deaths annually. Macrophages have been implicated in both the resolution and progression of the disease, but the drivers of these outcomes are poorly understood. We probed mouse lung transcriptomic datasets using the Digital Cell Quantifier algorithm to predict immune cell subsets that correlated with mild or severe influenza A virus (IAV) infection outcomes. We identified a unique lung macrophage population that transcriptionally resembled small serosal cavity macrophages and whose presence correlated with mild disease. Until now, the study of serosal macrophage translocation in the context of viral infections has been neglected. Here, we show that pleural macrophages (PMs) migrate from the pleural cavity to the lung after infection with IAV. We found that the depletion of PMs increased morbidity and pulmonary inflammation. There were increased proinflammatory cytokines in the pleural cavity and an influx of neutrophils within the lung. Our results show that PMs are recruited to the lung during IAV infection and contribute to recovery from influenza. This study expands our knowledge of PM plasticity and identifies a source of lung macrophages independent of monocyte recruitment and local proliferation.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Animales , Ratones , Humanos , Gripe Humana/genética , Pulmón , Macrófagos , Macrófagos Alveolares
2.
Nat Commun ; 12(1): 6790, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34815405

RESUMEN

Lineage commitment and differentiation is driven by the concerted action of master transcriptional regulators at their target chromatin sites. Multiple efforts have characterized the key transcription factors (TFs) that determine the various hematopoietic lineages. However, the temporal interactions between individual TFs and their chromatin targets during differentiation and how these interactions dictate lineage commitment remains poorly understood. Here we perform dense, daily, temporal profiling of chromatin accessibility (DNase I-seq) and gene expression changes (total RNA-seq) along ex vivo human erythropoiesis to comprehensively define developmentally regulated DNase I hypersensitive sites (DHSs) and transcripts. We link both distal DHSs to their target gene promoters and individual TFs to their target DHSs, revealing that the regulatory landscape is organized in distinct sequential regulatory modules that regulate lineage restriction and maturation. Finally, direct comparison of transcriptional dynamics (bulk and single-cell) and lineage potential between erythropoiesis and megakaryopoiesis uncovers differential fate commitment dynamics between the two lineages as they exit the stem and progenitor stage. Collectively, these data provide insights into the temporally regulated synergy of the cis- and the trans-regulatory components underlying hematopoietic lineage commitment and differentiation.


Asunto(s)
Linaje de la Célula/genética , Cromatina/genética , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/fisiología , Línea Celular , Cromatina/metabolismo , Ensayo de Unidades Formadoras de Colonias , Desoxirribonucleasa I/metabolismo , Humanos , Leucocitos Mononucleares , Cultivo Primario de Células , Regiones Promotoras Genéticas , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción/metabolismo
3.
Front Immunol ; 12: 681950, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34168651

RESUMEN

Dengue virus (DENV), a Flavivirus, causes a broad spectrum of disease in humans with key clinical signs including thrombocytopenia, vascular leakage and hemorrhaging. A major obstacle to understanding DENV immunity has been the lack of a validated immune-competent mouse model. Here, we report the infection profiles of human clinical isolates of DENV serotypes 1-4 in an immune-competent mouse model. We detected replicating DENV in the peritoneal cells, liver and the spleen that was generally resolved within 2 weeks. The DENV target cell types for infection were monocytes/macrophages, dendritic cells, endothelial cells, and we identified a novel DENV cellular target, fibroblast reticular cells of the spleen. We observed gross pathologies in the spleen and liver that are consistent with dengue disease, including hemorrhaging as well as transcriptional patterns suggesting that antiviral responses and tissue damage were induced. Key clinical blood parameters that define human DENV disease such as hemoconcentration, leukopenia and reduced number of platelets were also observed. Thus, immune-competent mice sustain replicating infection and experience signs, such as hemorrhaging, that define DENV disease in humans. This study thoroughly characterizes DENV1-4 infection in immune-competent mice and confirms the wild-type mouse model as a valid and reproducible system for investigating the mechanisms of DENV pathogenesis.


Asunto(s)
Virus del Dengue/inmunología , Dengue/inmunología , Dengue/virología , Interacciones Huésped-Patógeno/inmunología , Animales , Biopsia , Dengue/patología , Virus del Dengue/clasificación , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Regulación de la Expresión Génica , Huésped Inmunocomprometido , Ratones , Especificidad de Órganos , Serogrupo
4.
Nat Commun ; 10(1): 5101, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31704931

RESUMEN

Natural hosts of simian immunodeficiency virus (SIV) avoid AIDS despite lifelong infection. Here, we examined how this outcome is achieved by comparing a natural SIV host, African green monkey (AGM) to an AIDS susceptible species, rhesus macaque (RM). To asses gene expression profiles from acutely SIV infected AGMs and RMs, we developed a systems biology approach termed Conserved Gene Signature Analysis (CGSA), which compared RNA sequencing data from rectal AGM and RM tissues to various other species. We found that AGMs rapidly activate, and then maintain, evolutionarily conserved regenerative wound healing mechanisms in mucosal tissue. The wound healing protein fibronectin shows distinct tissue distribution and abundance kinetics in AGMs. Furthermore, AGM monocytes exhibit an embryonic development and repair/regeneration signature featuring TGF-ß and concomitant reduced expression of inflammatory genes compared to RMs. This regenerative wound healing process likely preserves mucosal integrity and prevents inflammatory insults that underlie immune exhaustion in RMs.


Asunto(s)
Fibronectinas/inmunología , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Factor de Crecimiento Transformador beta/inmunología , Cicatrización de Heridas/inmunología , Animales , Chlorocebus aethiops/genética , Chlorocebus aethiops/inmunología , Progresión de la Enfermedad , Fibronectinas/metabolismo , Mucosa Intestinal/metabolismo , Macaca mulatta/genética , Macaca mulatta/inmunología , Macrófagos/metabolismo , Recto/inmunología , Recto/metabolismo , Virus de la Inmunodeficiencia de los Simios , Biología de Sistemas , Transcriptoma , Factor de Crecimiento Transformador beta/genética , Cicatrización de Heridas/genética
5.
Vaccine ; 35(45): 6112-6121, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-28967519

RESUMEN

The host innate immune response to influenza virus is a key determinant of pathogenic outcomes and long-term protective immune responses against subsequent exposures. Here, we present a direct contrast of the host responses in primary differentiated human nasal epithelial cell (hNEC) cultures following infection with either a seasonal H3N2 influenza virus (WT) or the antigenically-matched live-attenuated vaccine (LAIV) strain. Comparison of the transcriptional profiles obtained 24 and 36h post-infection showed that the magnitude of gene expression was greater in LAIV infected relative to that observed in WT infected hNEC cultures. Functional enrichment analysis revealed that the antiviral and inflammatory responses were largely driven by type III IFN induction in both WT and LAIV infected cells. However, the enrichment of biological pathways involved in the recruitment of mononuclear leukocytes, antigen-presenting cells, and T lymphocytes was uniquely observed in LAIV infected cells. These observations were reflective of the host innate immune responses observed in individuals acutely infected with influenza viruses. These findings indicate that cell-intrinsic type III IFN-mediated innate immune responses in the nasal epithelium are not only crucial for viral clearance and attenuation, but may also play an important role in the induction of protective immune responses with live-attenuated vaccines.


Asunto(s)
Células Epiteliales/inmunología , Inmunidad Innata/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Mucosa Nasal/inmunología , Vacunas Atenuadas/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Línea Celular , Perros , Células Epiteliales/virología , Humanos , Subtipo H3N2 del Virus de la Influenza A/inmunología , Leucocitos Mononucleares , Células de Riñón Canino Madin Darby , Mucosa Nasal/virología , Linfocitos T/inmunología
6.
PLoS Pathog ; 13(5): e1006397, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28542576

RESUMEN

Fatal outcomes of Ebola virus (EBOV) infections are typically preceded by a 'sepsis-like' syndrome and lymphopenia despite T cells being resistant to Ebola infection. The mechanisms that lead to T lymphocytes death remain largely unknown; however, the degree of lymphopenia is highly correlative with fatalities. Here we investigated whether the addition of EBOV or its envelope glycoprotein (GP) to isolated primary human CD4+ T cells induced cell death. We observed a significant decrease in cell viability in a GP-dependent manner, which is suggestive of a direct role of GP in T cell death. Using immunoprecipitation assays and flow cytometry, we demonstrate that EBOV directly binds to CD4+ T cells through interaction of GP with TLR4. Transcriptome analysis revealed that the addition of EBOV to CD4+ T cells results in the significant upregulation of pathways associated with interferon signaling, pattern recognition receptors and intracellular activation of NFκB signaling pathway. Both transcriptome analysis and specific inhibitors allowed identification of apoptosis and necrosis as mechanisms associated with the observed T cell death following exposure to EBOV. The addition of the TLR4 inhibitor CLI-095 significantly reduced CD4+ T cell death induced by GP. EBOV stimulation of primary CD4+ T cells resulted in a significant increase in secreted TNFα; inhibition of TNFα-mediated signaling events significantly reduced T cell death while inhibitors of both necrosis and apoptosis similarly reduced EBOV-induced T cell death. Lastly, we show that stimulation with EBOV or GP augments monocyte maturation as determined by an overall increase in expression levels of markers of differentiation. Subsequently, the increased rates of cellular differentiation resulted in higher rates of infection further contributing to T cell death. These results demonstrate that GP directly subverts the host's immune response by increasing the susceptibility of monocytes to EBOV infection and triggering lymphopenia through direct and indirect mechanisms.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Ebolavirus/metabolismo , Fiebre Hemorrágica Ebola/fisiopatología , Proteínas del Envoltorio Viral/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Muerte Celular , Células Cultivadas , Ebolavirus/genética , Fiebre Hemorrágica Ebola/genética , Fiebre Hemorrágica Ebola/metabolismo , Fiebre Hemorrágica Ebola/virología , Interacciones Huésped-Patógeno , Humanos , Unión Proteica , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Proteínas del Envoltorio Viral/genética
7.
J Virol ; 91(11)2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28331091

RESUMEN

Ebola virus (EBOV) and Reston virus (RESTV) are members of the Ebolavirus genus which greatly differ in their pathogenicity. While EBOV causes a severe disease in humans characterized by a dysregulated inflammatory response and elevated cytokine and chemokine production, there are no reported disease-associated human cases of RESTV infection, suggesting that RESTV is nonpathogenic for humans. The underlying mechanisms determining the pathogenicity of different ebolavirus species are not yet known. In this study, we dissected the host response to EBOV and RESTV infection in primary human monocyte-derived macrophages (MDMs). As expected, EBOV infection led to a profound proinflammatory response, including strong induction of type I and type III interferons (IFNs). In contrast, RESTV-infected macrophages remained surprisingly silent. Early activation of IFN regulatory factor 3 (IRF3) and NF-κB was observed in EBOV-infected, but not in RESTV-infected, MDMs. In concordance with previous results, MDMs treated with inactivated EBOV and Ebola virus-like particles (VLPs) induced NF-κB activation mediated by Toll-like receptor 4 (TLR4) in a glycoprotein (GP)-dependent manner. This was not the case in cells exposed to live RESTV, inactivated RESTV, or VLPs containing RESTV GP, indicating that RESTV GP does not trigger TLR4 signaling. Our results suggest that the lack of immune activation in RESTV-infected MDMs contributes to lower pathogenicity by preventing the cytokine storm observed in EBOV infection. We further demonstrate that inhibition of TLR4 signaling abolishes EBOV GP-mediated NF-κB activation. This finding indicates that limiting the excessive TLR4-mediated proinflammatory response in EBOV infection should be considered as a potential supportive treatment option for EBOV disease.IMPORTANCE Emerging infectious diseases are a major public health concern, as exemplified by the recent devastating Ebola virus (EBOV) outbreak. Different ebolavirus species are associated with widely varying pathogenicity in humans, ranging from asymptomatic infections for Reston virus (RESTV) to severe disease with fatal outcomes for EBOV. In this comparative study of EBOV- and RESTV-infected human macrophages, we identified key differences in host cell responses. Consistent with previous data, EBOV infection is associated with a proinflammatory signature triggered by the surface glycoprotein (GP), which can be inhibited by blocking TLR4 signaling. In contrast, infection with RESTV failed to stimulate a strong host response in infected macrophages due to the inability of RESTV GP to stimulate TLR4. We propose that disparate proinflammatory host signatures contribute to the differences in pathogenicity reported for ebolavirus species and suggest that proinflammatory pathways represent an intriguing target for the development of novel therapeutics.


Asunto(s)
Ebolavirus/inmunología , Ebolavirus/patogenicidad , Interacciones Huésped-Patógeno , Macrófagos/virología , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Quimiocinas/inmunología , Quimiocinas/metabolismo , Chlorocebus aethiops , Citocinas/inmunología , Células Dendríticas/inmunología , Células Dendríticas/virología , Ebolavirus/fisiología , Perfilación de la Expresión Génica , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Interferones/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Células Vero , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA