Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Comput Biol Med ; 159: 106885, 2023 06.
Artículo en Inglés | MEDLINE | ID: covidwho-2290994

RESUMEN

Corona virus disease (COVID-19) has been emerged as pandemic infectious disease. The recent epidemiological data suggest that the smokers are more vulnerable to infection with COVID-19; however, the influence of smoking (SMK) on the COVID-19 infected patients and the mortality is not known yet. In this study, we aimed to discern the influence of SMK on COVID-19 infected patients utilizing the transcriptomics data of COVID-19 infected lung epithelial cells and transcriptomics data smoking matched with controls from lung epithelial cells. The bioinformatics based analysis revealed the molecular insights into the level of transcriptional changes and pathways which are important to identify the impact of smoking on COVID-19 infection and prevalence. We compared differentially expressed genes (DEGs) between COVID-19 and SMK and 59 DEGs were identified as consistently dysregulated at transcriptomics levels. The correlation network analyses were constructed for these common genes using WGCNA R package to see the relationship among these genes. Integration of DEGs with network analysis (protein-protein interaction) showed the presence of 9 hub proteins as key so called "candidate hub proteins" overlapped between COVID-19 patients and SMK. The Gene Ontology and pathways analysis demonstrated the enrichment of inflammatory pathway such as IL-17 signaling pathway, Interleukin-6 signaling, TNF signaling pathway and MAPK1/MAPK3 signaling pathways that might be the therapeutic targets in COVID-19 for smoking persons. The identified genes, pathways, hubs genes, and their regulators might be considered for establishment of key genes and drug targets for SMK and COVID-19.


Asunto(s)
COVID-19 , Humanos , COVID-19/genética , Transcriptoma/genética , SARS-CoV-2 , Pulmón , Células Epiteliales , Fumar/efectos adversos , Fumar/genética , Biología Computacional
2.
Sci Rep ; 12(1): 18168, 2022 Oct 28.
Artículo en Inglés | MEDLINE | ID: covidwho-2096749

RESUMEN

SARS-CoV-2 infection and disease severity are influenced by viral entry (VE) gene expression patterns in the airway epithelium. The similarities and differences of VE gene expression (ACE2, TMPRSS2, and CTSL) across nasal and bronchial compartments have not been fully characterized using matched samples from large cohorts. Gene expression data from 793 nasal and 1673 bronchial brushes obtained from individuals participating in lung cancer screening or diagnostic workup revealed that smoking status (current versus former) was the only clinical factor significantly and reproducibly associated with VE gene expression. The expression of ACE2 and TMPRSS2 was higher in smokers in the bronchus but not in the nose. scRNA-seq of nasal brushings indicated that ACE2 co-expressed genes were highly expressed in club and C15orf48+ secretory cells while TMPRSS2 co-expressed genes were highly expressed in keratinizing epithelial cells. In contrast, these ACE2 and TMPRSS2 modules were highly expressed in goblet cells in scRNA-seq from bronchial brushings. Cell-type deconvolution of the gene expression data confirmed that smoking increased the abundance of several secretory cell populations in the bronchus, but only goblet cells in the nose. The association of ACE2 and TMPRSS2 with smoking in the bronchus is due to their high expression in goblet cells which increase in abundance in current smoker airways. In contrast, in the nose, these genes are not predominantly expressed in cell populations modulated by smoking. In individuals with elevated lung cancer risk, smoking-induced VE gene expression changes in the nose likely have minimal impact on SARS-CoV-2 infection, but in the bronchus, smoking may lead to higher viral loads and more severe disease.


Asunto(s)
COVID-19 , Neoplasias Pulmonares , Humanos , SARS-CoV-2/genética , Enzima Convertidora de Angiotensina 2/genética , COVID-19/genética , Detección Precoz del Cáncer , Peptidil-Dipeptidasa A/metabolismo , Neoplasias Pulmonares/metabolismo , Bronquios/metabolismo , Fumar/efectos adversos , Fumar/genética
3.
Sci Rep ; 12(1): 1891, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: covidwho-1671627

RESUMEN

The COVID-19 pandemic has produced broad clinical manifestations, from asymptomatic infection to hospitalization and death. Despite progress from genomic and clinical epidemiology research, risk factors for developing severe COVID-19 are incompletely understood and identification of modifiable risk factors is desperately needed. We conducted linkage disequilibrium score regression (LDSR) analysis to estimate cross-trait genetic correlation between COVID-19 severity and various polygenic phenotypes. To attenuate the genetic contribution of smoking and BMI, we further conducted sensitivity analyses by pruning genomic regions associated with smoking/BMI and repeating LDSR analyses. We identified robust positive associations between the genetic architecture of severe COVID-19 and both BMI and smoking. We observed strong positive genetic correlation (rg) with diabetes (rg = 0.25) and shortness of breath walking on level ground (rg = 0.28) and novel protective associations with vitamin E (rg = - 0.53), calcium (rg = - 0.33), retinol (rg = - 0.59), Apolipoprotein A (rg = - 0.13), and HDL (rg = - 0.17), but no association with vitamin D (rg = - 0.02). Removing genomic regions associated with smoking and BMI generally attenuated the associations, but the associations with nutrient biomarkers persisted. This study provides a comprehensive assessment of the shared genetic architecture of COVID-19 severity and numerous clinical/physiologic parameters. Associations with blood and plasma-derived traits identified biomarkers for Mendelian randomization studies to explore causality and nominates therapeutic targets for clinical evaluation.


Asunto(s)
COVID-19/genética , Estudio de Asociación del Genoma Completo , Desequilibrio de Ligamiento/genética , Índice de Masa Corporal , COVID-19/etiología , Diabetes Mellitus/genética , Disnea/genética , Femenino , Humanos , Masculino , Análisis de la Aleatorización Mendeliana , Herencia Multifactorial , Nutrientes , Gravedad del Paciente , Fenotipo , Análisis de Regresión , Factores de Riesgo , Fumar/genética
4.
Epigenomics ; 14(3): 153-162, 2022 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1622527

RESUMEN

Smoking could predispose individuals to a more severe COVID-19 by upregulating a particular gene known as mdig, which is mediated through a number of well-known histone modifications. Smoking might regulate the transcription-activating H3K4me3 mark, along with the transcription-repressing H3K9me3 and H3K27me3 marks, in a way to favor SARS-CoV-2 entry by enhancing the expression of ACE2, NRP1 and NRP2, AT1R, CTSD and CTSL, PGE2 receptors 2-4, SLC6A20 and IL-6, all of which interact either directly or indirectly with important receptors, facilitating viral entry in COVID-19.


Lay abstract The role of smoking in development of several respiratory diseases has been clearly established. A significant proportion of these deleterious effects is mediated through epigenetic mechanisms, particularly histone modifications. Recent evidence indicates that smoking induces the expression of a mediator known as mdig, which in turn alters the transcription of several key proteins that have been implicated in development of COVID-19.


Asunto(s)
COVID-19/genética , Dioxigenasas/genética , Epigénesis Genética , Histona Demetilasas/genética , Histonas/genética , Proteínas Nucleares/genética , Procesamiento Proteico-Postraduccional , Fumar/genética , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/diagnóstico , COVID-19/metabolismo , COVID-19/virología , Catepsina D/genética , Catepsina D/metabolismo , Catepsina L/genética , Catepsina L/metabolismo , Dioxigenasas/metabolismo , Histona Demetilasas/metabolismo , Histonas/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Metilación , Neuropilina-1/genética , Neuropilina-1/metabolismo , Neuropilina-2/genética , Neuropilina-2/metabolismo , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo , Factores de Riesgo , SARS-CoV-2/genética , SARS-CoV-2/crecimiento & desarrollo , SARS-CoV-2/metabolismo , Fumar/metabolismo , Fumar/patología , Internalización del Virus
5.
Genome Med ; 13(1): 66, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: covidwho-1197350

RESUMEN

BACKGROUND: The large airway epithelial barrier provides one of the first lines of defense against respiratory viruses, including SARS-CoV-2 that causes COVID-19. Substantial inter-individual variability in individual disease courses is hypothesized to be partially mediated by the differential regulation of the genes that interact with the SARS-CoV-2 virus or are involved in the subsequent host response. Here, we comprehensively investigated non-genetic and genetic factors influencing COVID-19-relevant bronchial epithelial gene expression. METHODS: We analyzed RNA-sequencing data from bronchial epithelial brushings obtained from uninfected individuals. We related ACE2 gene expression to host and environmental factors in the SPIROMICS cohort of smokers with and without chronic obstructive pulmonary disease (COPD) and replicated these associations in two asthma cohorts, SARP and MAST. To identify airway biology beyond ACE2 binding that may contribute to increased susceptibility, we used gene set enrichment analyses to determine if gene expression changes indicative of a suppressed airway immune response observed early in SARS-CoV-2 infection are also observed in association with host factors. To identify host genetic variants affecting COVID-19 susceptibility in SPIROMICS, we performed expression quantitative trait (eQTL) mapping and investigated the phenotypic associations of the eQTL variants. RESULTS: We found that ACE2 expression was higher in relation to active smoking, obesity, and hypertension that are known risk factors of COVID-19 severity, while an association with interferon-related inflammation was driven by the truncated, non-binding ACE2 isoform. We discovered that expression patterns of a suppressed airway immune response to early SARS-CoV-2 infection, compared to other viruses, are similar to patterns associated with obesity, hypertension, and cardiovascular disease, which may thus contribute to a COVID-19-susceptible airway environment. eQTL mapping identified regulatory variants for genes implicated in COVID-19, some of which had pheWAS evidence for their potential role in respiratory infections. CONCLUSIONS: These data provide evidence that clinically relevant variation in the expression of COVID-19-related genes is associated with host factors, environmental exposures, and likely host genetic variation.


Asunto(s)
Bronquios , COVID-19/genética , Mucosa Respiratoria , SARS-CoV-2 , Adulto , Anciano , Anciano de 80 o más Años , Enzima Convertidora de Angiotensina 2/genética , Asma/genética , COVID-19/inmunología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/inmunología , Expresión Génica , Variación Genética , Humanos , Persona de Mediana Edad , Obesidad/genética , Obesidad/inmunología , Enfermedad Pulmonar Obstructiva Crónica/genética , Sitios de Carácter Cuantitativo , Factores de Riesgo , Fumar/genética
6.
Circulation ; 142(18): 1791-1793, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: covidwho-982724
7.
FEBS J ; 287(17): 3656-3663, 2020 09.
Artículo en Inglés | MEDLINE | ID: covidwho-960857

RESUMEN

There is an urgent need to address the devastating pandemic, COVID-19, caused by SARS-CoV-2. The efforts to understand the details of this disease in hope of providing effective treatments are commendable. It is clear now that the virus can cause far more damage in patients with comorbid conditions-particularly in those with respiratory, cardiovascular, or immune-compromised system-than in patients without such comorbidities. Drug use can further exacerbate the condition. In this regard, the ill effects of smoking are amply documented, and no doubt can be a confounding factor in COVID-19 progression. Although conflicting hypotheses on the potential role of nicotine in COVID-19 pathology have recently been offered, we believe that nicotine itself, through its interaction with the nicotinic cholinergic system, as well as ACE2, may not only be of use in a variety of neuropsychiatric and neurodegenerative diseases, but may also be of potential use in COVID-19. Thus, on one hand, while we strongly support smoking cessation as a means of harm reduction associated with COVID-19, on the other hand, we support a potential therapeutic role for nicotine, nicotinic agonists, or positive allosteric modulators of nicotinic cholinergic receptors in COVID-19, owing to their varied effects including mood regulation, anti-inflammatory, and purported interference with SARS-CoV-2 entry and/or replication.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , COVID-19/metabolismo , Síndrome de Liberación de Citoquinas/metabolismo , Nicotina/farmacología , Receptores Nicotínicos/genética , Fumar/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Enzima Convertidora de Angiotensina 2/inmunología , COVID-19/genética , COVID-19/inmunología , COVID-19/virología , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Receptores Nicotínicos/inmunología , Receptores Virales/genética , Receptores Virales/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Índice de Severidad de la Enfermedad , Transducción de Señal , Fumar/genética , Fumar/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología
9.
Int J Mol Sci ; 21(10)2020 May 21.
Artículo en Inglés | MEDLINE | ID: covidwho-327277

RESUMEN

The COVID-19 pandemic is marked by a wide range of clinical disease courses, ranging from asymptomatic to deadly. There have been many studies seeking to explore the correlations between COVID-19 clinical outcomes and various clinical variables, including age, sex, race, underlying medical problems, and social habits. In particular, the relationship between smoking and COVID-19 outcome is controversial, with multiple conflicting reports in the current literature. In this study, we aim to analyze how smoking may affect the SARS-CoV-2 infection rate. We analyzed sequencing data from lung and oral epithelial samples obtained from The Cancer Genome Atlas (TCGA). We found that the receptor and transmembrane protease necessary for SARS-CoV-2 entry into host cells, ACE2 and TMPRSS2, respectively, were upregulated in smoking samples from both lung and oral epithelial tissue. We then explored the mechanistic hypothesis that smoking may upregulate ACE2 expression through the upregulation of the androgen pathway. ACE2 and TMPRSS2 upregulation were both correlated to androgen pathway enrichment and the specific upregulation of central pathway regulatory genes. These data provide a potential model for the increased susceptibility of smoking patients to COVID-19 and encourage further exploration into the androgen and tobacco upregulation of ACE2 to understand the potential clinical ramifications.


Asunto(s)
Andrógenos/metabolismo , Infecciones por Coronavirus/metabolismo , Peptidil-Dipeptidasa A/genética , Neumonía Viral/metabolismo , Serina Endopeptidasas/genética , Fumar/metabolismo , Regulación hacia Arriba , Células Epiteliales Alveolares/metabolismo , Enzima Convertidora de Angiotensina 2 , COVID-19 , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/genética , Humanos , Mucosa Bucal/metabolismo , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/epidemiología , Neumonía Viral/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Serina Endopeptidasas/metabolismo , Fumar/epidemiología , Fumar/genética
10.
FEBS J ; 287(17): 3651-3655, 2020 09.
Artículo en Inglés | MEDLINE | ID: covidwho-10071

RESUMEN

The recent emergence of COVID-19 has resulted in a worldwide crisis, with large populations locked down and transportation links severed. While approximately 80% of infected individuals have minimal symptoms, around 15-20% need to be hospitalized, greatly stressing global healthcare systems. As of March 10, the death rate appears to be about 3.4%, although this number is highly stratified among different populations. Here, we focus on those individuals who have been exposed to nicotine prior to their exposure to the virus. We predict that these individuals are 'primed' to be at higher risk because nicotine can directly impact the putative receptor for the virus (ACE2) and lead to deleterious signaling in lung epithelial cells.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , COVID-19/epidemiología , Nicotina/efectos adversos , Pandemias , Receptores Nicotínicos/genética , Fumar/epidemiología , Glicoproteína de la Espiga del Coronavirus/genética , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/genética , COVID-19/fisiopatología , COVID-19/virología , Femenino , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Masculino , Receptores Nicotínicos/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , SARS-CoV-2/patogenicidad , Índice de Severidad de la Enfermedad , Factores Sexuales , Transducción de Señal , Fumar/genética , Fumar/fisiopatología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA