Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Nature ; 610(7931): 381-388, 2022 10.
Artículo en Inglés | MEDLINE | ID: covidwho-2050416

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and caused the devastating global pandemic of coronavirus disease 2019 (COVID-19), in part because of its ability to effectively suppress host cell responses1-3. In rare cases, viral proteins dampen antiviral responses by mimicking critical regions of human histone proteins4-8, particularly those containing post-translational modifications required for transcriptional regulation9-11. Recent work has demonstrated that SARS-CoV-2 markedly disrupts host cell epigenetic regulation12-14. However, how SARS-CoV-2 controls the host cell epigenome and whether it uses histone mimicry to do so remain unclear. Here we show that the SARS-CoV-2 protein encoded by ORF8 (ORF8) functions as a histone mimic of the ARKS motifs in histone H3 to disrupt host cell epigenetic regulation. ORF8 is associated with chromatin, disrupts regulation of critical histone post-translational modifications and promotes chromatin compaction. Deletion of either the ORF8 gene or the histone mimic site attenuates the ability of SARS-CoV-2 to disrupt host cell chromatin, affects the transcriptional response to infection and attenuates viral genome copy number. These findings demonstrate a new function of ORF8 and a mechanism through which SARS-CoV-2 disrupts host cell epigenetic regulation. Further, this work provides a molecular basis for the finding that SARS-CoV-2 lacking ORF8 is associated with decreased severity of COVID-19.


Asunto(s)
COVID-19 , Epigénesis Genética , Histonas , Interacciones Microbiota-Huesped , Imitación Molecular , SARS-CoV-2 , Proteínas Virales , COVID-19/genética , COVID-19/metabolismo , COVID-19/virología , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Epigenoma/genética , Histonas/química , Histonas/metabolismo , Humanos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo
2.
Biochem J ; 478(14): 2789-2791, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: covidwho-1526112

RESUMEN

Post-translational modifications (PTMs) on histone proteins are known as epigenetic marks that demarcate the status of chromatin. These modifications are 'read' by specific reader proteins, which in turn recruit additional factors to modulate chromatin accessibility and the activity of the underlying DNA. Accumulating evidence suggests that these modifications are not restricted solely to histones, many non-histone proteins may function in a similar way through mimicking the histones. In this commentary, we briefly discuss a systematic study of the discovery of histone H3 N-terminal mimicry proteins (H3TMs), and their implications in chromatin regulation and drug discoveries.


Asunto(s)
Cromatina/metabolismo , ADN/metabolismo , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Cromatina/genética , Ensamble y Desensamble de Cromatina , ADN/genética , Humanos , Lisina/metabolismo , Metilación , Modelos Biológicos
3.
Int J Mol Sci ; 22(19)2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1463708

RESUMEN

Social behavioral changes, including social isolation or loneliness, increase the risk for stress-related disorders, such as major depressive disorder, posttraumatic stress disorder (PTSD), and suicide, which share a strong neuroinflammatory etiopathogenetic component. The peroxisome-proliferator activated receptor (PPAR)-α, a newly discovered target involved in emotional behavior regulation, is a ligand-activated nuclear receptor and a transcription factor that, following stimulation by endogenous or synthetic ligands, may induce neuroprotective effects by modulating neuroinflammation, and improve anxiety and depression-like behaviors by enhancing neurosteroid biosynthesis. How stress affects epigenetic mechanisms with downstream effects on inflammation and emotional behavior remains poorly understood. We studied the effects of 4-week social isolation, using a mouse model of PTSD/suicide-like behavior, on hippocampal PPAR-α epigenetic modification. Decreased PPAR-α expression in the hippocampus of socially isolated mice was associated with increased levels of methylated cytosines of PPAR-α gene CpG-rich fragments and deficient neurosteroid biosynthesis. This effect was associated with increased histone deacetylases (HDAC)1, methyl-cytosine binding protein (MeCP)2 and decreased ten-eleven translocator (TET)2 expression, which favor hypermethylation. These alterations were associated with increased TLR-4 and pro-inflammatory markers (e.g., TNF-α,), mediated by NF-κB signaling in the hippocampus of aggressive mice. This study contributes the first evidence of stress-induced brain PPAR-α epigenetic regulation. Social isolation stress may constitute a risk factor for inflammatory-based psychiatric disorders associated with neurosteroid deficits, and targeting epigenetic marks linked to PPAR-α downregulation may offer a valid therapeutic approach.


Asunto(s)
Agresión , Hipocampo/metabolismo , Inflamación/etiología , PPAR alfa/genética , Aislamiento Social , Estrés Psicológico , Agresión/psicología , Animales , Conducta Animal , Ensamble y Desensamble de Cromatina , Islas de CpG , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Epigénesis Genética , Expresión Génica , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Metilación , Ratones , PPAR alfa/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal
4.
Nat Cell Biol ; 23(6): 620-630, 2021 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1263492

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection often causes severe complications and even death. However, asymptomatic infection has also been reported, highlighting the difference in immune responses among individuals. Here we performed single-cell chromatin accessibility and T cell-receptor analyses of peripheral blood mononuclear cells collected from individuals convalescing from COVID-19 and healthy donors. Chromatin remodelling was observed in both innate and adaptive immune cells in the individuals convalescing from COVID-19. Compared with healthy donors, recovered individuals contained abundant TBET-enriched CD16+ and IRF1-enriched CD14+ monocytes with sequential trained and activated epigenomic states. The B-cell lineage in recovered individuals exhibited an accelerated developmental programme from immature B cells to antibody-producing plasma cells. Finally, an integrated analysis of single-cell T cell-receptor clonality with the chromatin accessibility landscape revealed the expansion of putative SARS-CoV-2-specific CD8+ T cells with epigenomic profiles that promote the differentiation of effector or memory cells. Overall, our data suggest that immune cells of individuals convalescing from COVID-19 exhibit global remodelling of the chromatin accessibility landscape, indicative of the establishment of immunological memory.


Asunto(s)
COVID-19/genética , Epigénesis Genética , Epigenómica , Genes Codificadores de los Receptores de Linfocitos T , Memoria Inmunológica , Subgrupos Linfocitarios/inmunología , Monocitos/inmunología , SARS-CoV-2/inmunología , Análisis de la Célula Individual , Inmunidad Adaptativa , Adolescente , Adulto , Anciano , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/virología , Estudios de Casos y Controles , Diferenciación Celular , Ensamble y Desensamble de Cromatina , Femenino , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Subgrupos Linfocitarios/metabolismo , Subgrupos Linfocitarios/virología , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Monocitos/virología , SARS-CoV-2/patogenicidad , Adulto Joven
5.
Signal Transduct Target Ther ; 5(1): 125, 2020 07 13.
Artículo en Inglés | MEDLINE | ID: covidwho-654479

RESUMEN

Stress proteins (SPs) including heat-shock proteins (HSPs), RNA chaperones, and ER associated stress proteins are molecular chaperones essential for cellular homeostasis. The major functions of HSPs include chaperoning misfolded or unfolded polypeptides, protecting cells from toxic stress, and presenting immune and inflammatory cytokines. Regarded as a double-edged sword, HSPs also cooperate with numerous viruses and cancer cells to promote their survival. RNA chaperones are a group of heterogeneous nuclear ribonucleoproteins (hnRNPs), which are essential factors for manipulating both the functions and metabolisms of pre-mRNAs/hnRNAs transcribed by RNA polymerase II. hnRNPs involve in a large number of cellular processes, including chromatin remodelling, transcription regulation, RNP assembly and stabilization, RNA export, virus replication, histone-like nucleoid structuring, and even intracellular immunity. Dysregulation of stress proteins is associated with many human diseases including human cancer, cardiovascular diseases, neurodegenerative diseases (e.g., Parkinson's diseases, Alzheimer disease), stroke and infectious diseases. In this review, we summarized the biologic function of stress proteins, and current progress on their mechanisms related to virus reproduction and diseases caused by virus infections. As SPs also attract a great interest as potential antiviral targets (e.g., COVID-19), we also discuss the present progress and challenges in this area of HSP-based drug development, as well as with compounds already under clinical evaluation.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Proteínas de Choque Térmico/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Interacciones Huésped-Patógeno/efectos de los fármacos , Neumonía Viral/tratamiento farmacológico , Antivirales/síntesis química , Betacoronavirus/genética , Betacoronavirus/patogenicidad , COVID-19 , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Regulación de la Expresión Génica , Proteínas de Choque Térmico/agonistas , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Choque Térmico/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/agonistas , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Terapia Molecular Dirigida/métodos , Pandemias , Neumonía Viral/genética , Neumonía Viral/patología , Neumonía Viral/virología , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Transducción de Señal , Transcripción Genética/efectos de los fármacos , Replicación Viral/efectos de los fármacos
6.
Protein Cell ; 11(10): 740-770, 2020 10.
Artículo en Inglés | MEDLINE | ID: covidwho-709445

RESUMEN

Age-associated changes in immune cells have been linked to an increased risk for infection. However, a global and detailed characterization of the changes that human circulating immune cells undergo with age is lacking. Here, we combined scRNA-seq, mass cytometry and scATAC-seq to compare immune cell types in peripheral blood collected from young and old subjects and patients with COVID-19. We found that the immune cell landscape was reprogrammed with age and was characterized by T cell polarization from naive and memory cells to effector, cytotoxic, exhausted and regulatory cells, along with increased late natural killer cells, age-associated B cells, inflammatory monocytes and age-associated dendritic cells. In addition, the expression of genes, which were implicated in coronavirus susceptibility, was upregulated in a cell subtype-specific manner with age. Notably, COVID-19 promoted age-induced immune cell polarization and gene expression related to inflammation and cellular senescence. Therefore, these findings suggest that a dysregulated immune system and increased gene expression associated with SARS-CoV-2 susceptibility may at least partially account for COVID-19 vulnerability in the elderly.


Asunto(s)
Envejecimiento/inmunología , Betacoronavirus , Infecciones por Coronavirus/inmunología , Sistema Inmunológico/inmunología , Pandemias , Neumonía Viral/inmunología , Análisis de la Célula Individual , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Linfocitos T CD4-Positivos/metabolismo , COVID-19 , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Síndrome de Liberación de Citoquinas/etiología , Síndrome de Liberación de Citoquinas/inmunología , Citocinas/biosíntesis , Citocinas/genética , Susceptibilidad a Enfermedades , Citometría de Flujo/métodos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Reordenamiento Génico , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/crecimiento & desarrollo , Inmunocompetencia/genética , Inflamación/genética , Inflamación/inmunología , Espectrometría de Masas/métodos , Persona de Mediana Edad , SARS-CoV-2 , Análisis de Secuencia de ARN , Transcriptoma , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA