Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell Rep Med ; 4(5): 101024, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: covidwho-2295352

RESUMEN

RNA viruses continue to remain a threat for potential pandemics due to their rapid evolution. Potentiating host antiviral pathways to prevent or limit viral infections is a promising strategy. Thus, by testing a library of innate immune agonists targeting pathogen recognition receptors, we observe that Toll-like receptor 3 (TLR3), stimulator of interferon genes (STING), TLR8, and Dectin-1 ligands inhibit arboviruses, Chikungunya virus (CHIKV), West Nile virus, and Zika virus to varying degrees. STING agonists (cAIMP, diABZI, and 2',3'-cGAMP) and Dectin-1 agonist scleroglucan demonstrate the most potent, broad-spectrum antiviral function. Furthermore, STING agonists inhibit severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and enterovirus-D68 (EV-D68) infection in cardiomyocytes. Transcriptome analysis reveals that cAIMP treatment rescue cells from CHIKV-induced dysregulation of cell repair, immune, and metabolic pathways. In addition, cAIMP provides protection against CHIKV in a chronic CHIKV-arthritis mouse model. Our study describes innate immune signaling circuits crucial for RNA virus replication and identifies broad-spectrum antivirals effective against multiple families of pandemic potential RNA viruses.


Asunto(s)
COVID-19 , Virus Chikungunya , Virus ARN , Infección por el Virus Zika , Virus Zika , Animales , Ratones , SARS-CoV-2 , Antivirales/farmacología , Antivirales/uso terapéutico , Virus Chikungunya/fisiología , Inmunidad Innata
3.
Microb Genom ; 9(3)2023 03.
Artículo en Inglés | MEDLINE | ID: covidwho-2277598

RESUMEN

Novel variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continue to emerge as the coronavirus disease 2019 (COVID-19) pandemic extends into its fourth year. Understanding SARS-CoV-2 circulation in university populations is vital for effective interventions in higher education settings and will inform public health policy during pandemics. In this study, we performed whole-genome sequencing of 537 of 1717 SARS-CoV-2-positive nasopharyngeal/nasal swab samples collected over a nearly 20-month period from two university populations in Wisconsin, USA. We observed that the viral sequences were distributed into 57 lineages/sub-lineages belonging to 15 clades, of which the majority were from 21K (omicron, 36.13 %) and 21J (delta, 30.91 %). Nearly 40 % (213) of the sequences were omicron, of which BA.1 and its eight descendent lineages accounted for 91 %, while the remaining belonged to BA.2 and its six descendent lineages. Independent analysis of the sequences from these two universities revealed significant differences in the circulating SARS-CoV-2 variants. Phylogenetic analysis of university sequences with a global sub-dataset demonstrated that the sequences of the same lineages from the university populations were more closely related. Genome-based analysis of closely related strains, along with phylogenetic clusters and mutational differences, identified that potential virus transmission occurred within and between universities, as well as between the university and the local community. Although this study improves our understanding of the distinct transmission patterns of circulating variants in local universities, expanding genomic surveillance capacity will aid local jurisdictions not only in identifying emerging SARS-CoV-2 variants, but also in improving data-driven public health mitigation and policy efforts.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Universidades , Wisconsin/epidemiología , Filogenia , COVID-19/epidemiología , Genómica
4.
PLoS Biol ; 20(11): e3001851, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: covidwho-2109273

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), responsible for the Coronavirus Disease 2019 (COVID-19) pandemic, causes respiratory failure and damage to multiple organ systems. The emergence of viral variants poses a risk of vaccine failures and prolongation of the pandemic. However, our understanding of the molecular basis of SARS-CoV-2 infection and subsequent COVID-19 pathophysiology is limited. In this study, we have uncovered a critical role for the evolutionarily conserved Hippo signaling pathway in COVID-19 pathogenesis. Given the complexity of COVID-19-associated cell injury and immunopathogenesis processes, we investigated Hippo pathway dynamics in SARS-CoV-2 infection by utilizing COVID-19 lung samples and human cell models based on pluripotent stem cell-derived cardiomyocytes (PSC-CMs) and human primary lung air-liquid interface (ALI) cultures. SARS-CoV-2 infection caused activation of the Hippo signaling pathway in COVID-19 lung and in vitro cultures. Both parental and Delta variant of concern (VOC) strains induced Hippo pathway. The chemical inhibition and gene knockdown of upstream kinases MST1/2 and LATS1 resulted in significantly enhanced SARS-CoV-2 replication, indicating antiviral roles. Verteporfin, a pharmacological inhibitor of the Hippo pathway downstream transactivator, YAP, significantly reduced virus replication. These results delineate a direct antiviral role for Hippo signaling in SARS-CoV-2 infection and the potential for this pathway to be pharmacologically targeted to treat COVID-19.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Vía de Señalización Hippo , Antivirales/farmacología
5.
Viruses ; 14(11)2022 Nov 06.
Artículo en Inglés | MEDLINE | ID: covidwho-2099866

RESUMEN

New variants of SARS-CoV-2 continue to evolve. The novel SARS-CoV-2 variant of concern (VOC) B.1.1.529 (Omicron) was particularly menacing due to the presence of numerous consequential mutations. In this study, we reviewed about 12 million SARS-CoV-2 genomic and associated metadata using extensive bioinformatic approaches to understand how evolutionary and mutational changes affect Omicron variant properties. Subsampled global data based analysis of molecular clock in the phylogenetic tree showed 29.56 substitutions per year as the evolutionary rate of five VOCs. We observed extensive mutational changes in the spike structural protein of the Omicron variant. A total of 20% of 7230 amino acid and structural changes exclusive to Omicron's spike protein were detected in the receptor binding domain (RBD), suggesting differential selection pressures exerted during evolution. Analyzing key drug targets revealed mutation-derived differential binding affinities between Delta and Omicron variants. Nine single-RBD substitutions were detected within the binding site of approved therapeutic monoclonal antibodies. T-cell epitope prediction revealed eight immunologically important functional hotspots in three conserved non-structural proteins. A universal vaccine based on these regions may likely protect against all these SARS-CoV-2 variants. We observed key structural changes in the spike protein, which decreased binding affinities, indicating that these changes may help the virus escape host cellular immunity. These findings emphasize the need for continuous genomic surveillance of SARS-CoV-2 to better understand how novel mutations may impact viral spread and disease outcome.


Asunto(s)
Antivirales , COVID-19 , Evasión Inmune , SARS-CoV-2 , Humanos , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/inmunología , COVID-19/virología , Mutación , Filogenia , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Proteínas del Envoltorio Viral/genética
6.
FASEB journal : official publication of the Federation of American Societies for Experimental Biology ; 36(Suppl 1), 2022.
Artículo en Inglés | EuropePMC | ID: covidwho-1970253

RESUMEN

SARS‐CoV‐2 is responsible for the ongoing COVID‐19 pandemic, which causes respiratory failure and damage to multiple organ systems. Emergence of new variants of concern (VOCs), including Omicron can potentially render the current vaccines ineffective. However, our understanding of COVID‐19 pathophysiology and molecular basis of SARS‐CoV‐2 infection is very limited. The role of the Hippo signaling pathway, an evolutionarily conserved organogenesis circuitry, in tissue inflammation and innate immune response is beginning to be understood. Given the complexity of COVID‐19 associated cell injury and immunopathogenesis processes, we investigated this Hippo pathway dynamic in SARS‐CoV‐2 infection by utilizing COVID‐19 lung samples, transcriptome and human cell models based on pluripotent stem cell‐derived cardiomyocytes (PSC‐CMs) and human primary lung air‐liquid interface (ALI) culture. The SARS‐CoV‐2 infection resulted in stoppage of cardiomyocyte beating and extensive apoptotic cell death. Especially the infection caused activation of Hippo signaling pathway in cardiomyocytes, as shown by increased level of phosphorylated form of YAP, a downstream transcriptional co‐factor involved in tissue growth, mitochondrial biogenesis and innate immunity. Similar activation was noted in SARS‐CoV‐2 infected lung ALI epithelial cells and COVID‐19 lung autopsy samples. The shRNA‐mediated partial knockdown and pharmacological inhibitor of YAP/TAZ resulted in significantly reduced SARS‐CoV‐2 replication, whereas inhibition of Hippo pathway upstream LATS1 and MST1 kinases led to enhanced virus replication. These results indicate a direct role of Hippo signaling in SARS‐CoV‐2 mediated disease pathogenesis and this pathway can be pharmacologically targeted to treat COVID‐19.

7.
J Immunol ; 208(1): 74-84, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1534334

RESUMEN

ORAI1 and stromal interaction molecule 1 (STIM1) are the critical mediators of store-operated Ca2+ entry by acting as the pore subunit and an endoplasmic reticulum-resident signaling molecule, respectively. In addition to Ca2+ signaling, STIM1 is also involved in regulation of the type I IFN (IFN-I) response. To examine their potential role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, we generated ORAI1 and STIM1 knockout human HEK293-angiotensin-converting enzyme 2 cells and checked their responses. STIM1 knockout cells showed strong resistance to SARS-CoV-2 infection as a result of enhanced IFN-I response. On the contrary, ORAI1 deletion induced high susceptibility to SARS-CoV-2 infection. Mechanistically, ORAI1 knockout cells showed reduced homeostatic cytoplasmic Ca2+ concentration and severe impairment in tonic IFN-I signaling. Transcriptome analysis showed downregulation of multiple antiviral signaling pathways in ORAI1 knockout cells, likely because of reduced expression of the Ca2+-dependent transcription factors of the AP-1 family and MEF2C Accordingly, modulation of homeostatic Ca2+ concentration by pretreatment with ORAI1 blocker or agonist could influence baseline IFNB expression and resistance to SARS-CoV-2 infection in a human lung epithelial cell line. Our results identify a novel role of ORAI1-mediated Ca2+ signaling in regulating the tonic IFN-I levels, which determine host resistance to SARS-CoV-2 infection.


Asunto(s)
COVID-19/metabolismo , Interferón Tipo I/metabolismo , Pulmón/inmunología , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Mucosa Respiratoria/metabolismo , SARS-CoV-2/fisiología , Molécula de Interacción Estromal 1/metabolismo , Células A549 , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/inmunología , Señalización del Calcio , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Resistencia a la Enfermedad , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Pulmón/virología , Factores de Transcripción MEF2/genética , Proteínas de Neoplasias/genética , Proteína ORAI1/genética , Molécula de Interacción Estromal 1/genética , Factor de Transcripción AP-1/genética
8.
FEBS Lett ; 595(23): 2854-2871, 2021 12.
Artículo en Inglés | MEDLINE | ID: covidwho-1508599

RESUMEN

SARS-CoV-2 has infected hundreds of millions of people with over four million dead, resulting in one of the worst global pandemics in recent history. Neurological symptoms associated with COVID-19 include anosmia, ageusia, headaches, confusion, delirium, and strokes. These may manifest due to viral entry into the central nervous system (CNS) through the blood-brain barrier (BBB) by means of ill-defined mechanisms. Here, we summarize the abilities of SARS-CoV-2 and other neurotropic RNA viruses, including Zika virus and Nipah virus, to cross the BBB into the CNS, highlighting the role of magnetic resonance imaging (MRI) in assessing presence and severity of brain structural changes in COVID-19 patients. We present new insight into key mutations in SARS-CoV-2 variants B.1.1.7 (P681H) and B.1.617.2 (P681R), which may impact on neuropilin 1 (NRP1) binding and CNS invasion. We postulate that SARS-CoV-2 may infect both peripheral cells capable of crossing the BBB and brain endothelial cells to traverse the BBB and spread into the brain. COVID-19 patients can be followed up with MRI modalities to better understand the long-term effects of COVID-19 on the brain.


Asunto(s)
Barrera Hematoencefálica , Infecciones por Henipavirus , Virus Nipah , SARS-CoV-2 , Infección por el Virus Zika , Virus Zika , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Barrera Hematoencefálica/virología , COVID-19/epidemiología , COVID-19/genética , COVID-19/metabolismo , COVID-19/fisiopatología , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/genética , Infecciones por Henipavirus/metabolismo , Infecciones por Henipavirus/fisiopatología , Humanos , Mutación , Virus Nipah/genética , Virus Nipah/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Virus Zika/genética , Virus Zika/metabolismo , Infección por el Virus Zika/epidemiología , Infección por el Virus Zika/genética , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/fisiopatología
9.
Cell Rep ; 35(1): 108940, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1157178

RESUMEN

SARS-CoV-2 has currently precipitated the COVID-19 global health crisis. We developed a medium-throughput drug-screening system and identified a small-molecule library of 34 of 430 protein kinase inhibitors that were capable of inhibiting the SARS-CoV-2 cytopathic effect in human epithelial cells. These drug inhibitors are in various stages of clinical trials. We detected key proteins involved in cellular signaling pathways mTOR-PI3K-AKT, ABL-BCR/MAPK, and DNA-damage response that are critical for SARS-CoV-2 infection. A drug-protein interaction-based secondary screen confirmed compounds, such as the ATR kinase inhibitor berzosertib and torin2 with anti-SARS-CoV-2 activity. Berzosertib exhibited potent antiviral activity against SARS-CoV-2 in multiple cell types and blocked replication at the post-entry step. Berzosertib inhibited replication of SARS-CoV-1 and the Middle East respiratory syndrome coronavirus (MERS-CoV) as well. Our study highlights key promising kinase inhibitors to constrain coronavirus replication as a host-directed therapy in the treatment of COVID-19 and beyond as well as provides an important mechanism of host-pathogen interactions.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Daño del ADN , Isoxazoles/farmacología , Pirazinas/farmacología , SARS-CoV-2/fisiología , Replicación Viral/efectos de los fármacos , Células A549 , Animales , COVID-19/metabolismo , COVID-19/patología , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Células HEK293 , Células HeLa , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Coronavirus del Síndrome Respiratorio de Oriente Medio/metabolismo , Células Vero
10.
Nat Commun ; 12(1): 1876, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: covidwho-1152854

RESUMEN

Viruses hijack host cell metabolism to acquire the building blocks required for replication. Understanding how SARS-CoV-2 alters host cell metabolism may lead to potential treatments for COVID-19. Here we profile metabolic changes conferred by SARS-CoV-2 infection in kidney epithelial cells and lung air-liquid interface (ALI) cultures, and show that SARS-CoV-2 infection increases glucose carbon entry into the TCA cycle via increased pyruvate carboxylase expression. SARS-CoV-2 also reduces oxidative glutamine metabolism while maintaining reductive carboxylation. Consistent with these changes, SARS-CoV-2 infection increases the activity of mTORC1 in cell lines and lung ALI cultures. Lastly, we show evidence of mTORC1 activation in COVID-19 patient lung tissue, and that mTORC1 inhibitors reduce viral replication in kidney epithelial cells and lung ALI cultures. Our results suggest that targeting mTORC1 may be a feasible treatment strategy for COVID-19 patients, although further studies are required to determine the mechanism of inhibition and potential efficacy in patients.


Asunto(s)
COVID-19/patología , Ciclo del Ácido Cítrico/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Benzamidas/farmacología , Línea Celular , Chlorocebus aethiops , Glucosa/metabolismo , Glutamina/metabolismo , Células HEK293 , Humanos , Pulmón/metabolismo , Pulmón/virología , Morfolinas/farmacología , Naftiridinas/farmacología , Pirimidinas/farmacología , Piruvato Carboxilasa/biosíntesis , SARS-CoV-2/metabolismo , Células Vero , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA