Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Nature ; 609(7928): 754-760, 2022 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1984401

RESUMEN

Identifying the host genetic factors underlying severe COVID-19 is an emerging challenge1-5. Here we conducted a genome-wide association study (GWAS) involving 2,393 cases of COVID-19 in a cohort of Japanese individuals collected during the initial waves of the pandemic, with 3,289 unaffected controls. We identified a variant on chromosome 5 at 5q35 (rs60200309-A), close to the dedicator of cytokinesis 2 gene (DOCK2), which was associated with severe COVID-19 in patients less than 65 years of age. This risk allele was prevalent in East Asian individuals but rare in Europeans, highlighting the value of genome-wide association studies in non-European populations. RNA-sequencing analysis of 473 bulk peripheral blood samples identified decreased expression of DOCK2 associated with the risk allele in these younger patients. DOCK2 expression was suppressed in patients with severe cases of COVID-19. Single-cell RNA-sequencing analysis (n = 61 individuals) identified cell-type-specific downregulation of DOCK2 and a COVID-19-specific decreasing effect of the risk allele on DOCK2 expression in non-classical monocytes. Immunohistochemistry of lung specimens from patients with severe COVID-19 pneumonia showed suppressed DOCK2 expression. Moreover, inhibition of DOCK2 function with CPYPP increased the severity of pneumonia in a Syrian hamster model of SARS-CoV-2 infection, characterized by weight loss, lung oedema, enhanced viral loads, impaired macrophage recruitment and dysregulated type I interferon responses. We conclude that DOCK2 has an important role in the host immune response to SARS-CoV-2 infection and the development of severe COVID-19, and could be further explored as a potential biomarker and/or therapeutic target.


Asunto(s)
COVID-19 , Proteínas Activadoras de GTPasa , Estudio de Asociación del Genoma Completo , Factores de Intercambio de Guanina Nucleótido , Interacciones Microbiota-Huesped , SARS-CoV-2 , Alelos , Animales , COVID-19/complicaciones , COVID-19/genética , COVID-19/inmunología , COVID-19/fisiopatología , Modelos Animales de Enfermedad , Proteínas Activadoras de GTPasa/antagonistas & inhibidores , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Predisposición Genética a la Enfermedad , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Japón , Pulmón/patología , Macrófagos , Mesocricetus , Persona de Mediana Edad , Neumonía/complicaciones , Pirazoles/farmacología , RNA-Seq , SARS-CoV-2/patogenicidad , Carga Viral , Pérdida de Peso
2.
JCI Insight ; 7(11)2022 06 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1807764

RESUMEN

COVID-19 infection causes collapse of glomerular capillaries and loss of podocytes, culminating in a severe kidney disease called COVID-19-associated nephropathy (COVAN). The underlying mechanism of COVAN is unknown. We hypothesized that cytokines induced by COVID-19 trigger expression of pathogenic APOL1 via JAK/STAT signaling, resulting in podocyte loss and COVAN phenotype. Here, based on 9 biopsy-proven COVAN cases, we demonstrated for the first time, to the best of our knowledge, that APOL1 protein was abundantly expressed in podocytes and glomerular endothelial cells (GECs) of COVAN kidneys but not in controls. Moreover, a majority of patients with COVAN carried 2 APOL1 risk alleles. We show that recombinant cytokines induced by SARS-CoV-2 acted synergistically to drive APOL1 expression through the JAK/STAT pathway in primary human podocytes, GECs, and kidney micro-organoids derived from a carrier of 2 APOL1 risk alleles, but expression was blocked by a JAK1/2 inhibitor, baricitinib. We demonstrate that cytokine-induced JAK/STAT/APOL1 signaling reduced the viability of kidney organoid podocytes but was rescued by baricitinib. Together, our results support the conclusion that COVID-19-induced cytokines are sufficient to drive COVAN-associated podocytopathy via JAK/STAT/APOL1 signaling and that JAK inhibitors could block this pathogenic process. These findings suggest JAK inhibitors may have therapeutic benefits for managing cytokine-induced, APOL1-mediated podocytopathy.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Citocinas , Inhibidores de las Cinasas Janus , Enfermedades Renales , Apolipoproteína L1/genética , Azetidinas/farmacología , COVID-19/metabolismo , Citocinas/metabolismo , Células Endoteliales/metabolismo , Humanos , Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/metabolismo , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Enfermedades Renales/virología , Organoides/metabolismo , Purinas/farmacología , Pirazoles/farmacología , SARS-CoV-2/aislamiento & purificación , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
3.
Clin Pharmacol Ther ; 111(6): 1324-1333, 2022 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1802136

RESUMEN

Cystic fibrosis transmembrane conductance regulator (CFTR) modulating therapies, including elexacaftor-tezacaftor-ivacaftor, are primarily eliminated through cytochrome P450 (CYP) 3A-mediated metabolism. This creates a therapeutic challenge to the treatment of coronavirus disease 2019 (COVID-19) with nirmatrelvir-ritonavir in people with cystic fibrosis (CF) due to the potential for significant drug-drug interactions (DDIs). However, the population with CF is more at risk of serious illness following COVID-19 infection and hence it is important to manage the DDI risk and provide treatment options. CYP3A-mediated DDI of elexacaftor-tezacaftor-ivacaftor was evaluated using a physiologically-based pharmacokinetic modeling approach. Modeling was performed incorporating physiological information and drug-dependent parameters of elexacaftor-tezacaftor-ivacaftor to predict the effect of ritonavir (the CYP3A inhibiting component of the combination) on the pharmacokinetics of elexacaftor-tezacaftor-ivacaftor. The elexacaftor-tezacaftor-ivacaftor models were verified using independent clinical pharmacokinetic and DDI data of elexacaftor-tezacaftor-ivacaftor with a range of CYP3A modulators. When ritonavir was administered on Days 1 through 5, the predicted area under the curve (AUC) ratio of ivacaftor (the most sensitive CYP3A substrate) on Day 6 was 9.31, indicating that its metabolism was strongly inhibited. Based on the predicted DDI, the dose of elexacaftor-tezacaftor-ivacaftor should be reduced when coadministered with nirmatrelvir-ritonavir to elexacaftor 200 mg-tezacaftor 100 mg-ivacaftor 150 mg on Days 1 and 5, with delayed resumption of full-dose elexacaftor-tezacaftor-ivacaftor on Day 9, considering the residual inhibitory effect of ritonavir as a mechanism-based inhibitor. The simulation predicts a regimen of elexacaftor-tezacaftor-ivacaftor administered concomitantly with nirmatrelvir-ritonavir in people with CF that will likely decrease the impact of the drug interaction.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Fibrosis Quística , Aminofenoles/farmacología , Benzodioxoles/farmacología , Agonistas de los Canales de Cloruro/uso terapéutico , Fibrosis Quística/tratamiento farmacológico , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Citocromo P-450 CYP3A/metabolismo , Combinación de Medicamentos , Interacciones Farmacológicas , Humanos , Indoles/farmacología , Lactamas/farmacocinética , Leucina/farmacocinética , Mutación , Nitrilos/farmacocinética , Prolina/farmacocinética , Pirazoles/farmacología , Piridinas/farmacología , Pirrolidinas , Quinolinas/farmacología , Quinolonas , Ritonavir/farmacocinética
4.
Mol Cell Biochem ; 477(3): 711-726, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: covidwho-1616202

RESUMEN

The novel coronavirus pandemic has emerged as one of the significant medical-health challenges of the current century. The World Health Organization has named this new virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since the first detection of SARS-CoV-2 in November 2019 in Wuhan, China, physicians, researchers, and others have made it their top priority to find drugs and cures that can effectively treat patients and reduce mortality rates. The symptoms of Coronavirus Disease 2019 (COVID-19) include fever, dry cough, body aches, and anosmia. Various therapeutic compounds have been investigated and applied to mitigate the symptoms in COVID-19 patients and cure the disease. Degenerative virus analyses of the infection incidence and COVID-19 have demonstrated that SARS-CoV-2 penetrates the pulmonary alveoli's endothelial cells through Angiotensin-Converting Enzyme 2 (ACE2) receptors on the membrane, stimulates various signaling pathways and causes excessive secretion of cytokines. The continuous triggering of the innate and acquired immune system, as well as the overproduction of pro-inflammatory factors, cause a severe condition in the COVID-19 patients, which is called "cytokine storm". It can lead to acute respiratory distress syndrome (ARDS) in critical patients. Severe and critical COVID-19 cases demand oxygen therapy and mechanical ventilator support. Various drugs, including immunomodulatory and immunosuppressive agents (e.g., monoclonal antibodies (mAbs) and interleukin antagonists) have been utilized in clinical trials. However, the studies and clinical trials have documented diverging findings, which seem to be due to the differences in these drugs' possible mechanisms of action. These drugs' mechanism of action generally includes suppressing or modulating the immune system, preventing the development of cytokine storm via various signaling pathways, and enhancing the blood vessels' diameter in the lungs. In this review article, multiple medications from different drug families are discussed, and their possible mechanisms of action are also described.


Asunto(s)
Antivirales/inmunología , Tratamiento Farmacológico de COVID-19 , Agentes Inmunomoduladores/farmacología , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Antivirales/farmacología , Azetidinas/inmunología , Azetidinas/farmacología , COVID-19/etiología , Dexametasona/inmunología , Dexametasona/farmacología , Famotidina/inmunología , Famotidina/farmacología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/inmunología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Infliximab/inmunología , Infliximab/farmacología , Proteína Antagonista del Receptor de Interleucina 1/inmunología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Melatonina/inmunología , Melatonina/farmacología , Purinas/inmunología , Purinas/farmacología , Pirazoles/inmunología , Pirazoles/farmacología , Sulfonamidas/inmunología , Sulfonamidas/farmacología
5.
Int Immunopharmacol ; 103: 108463, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: covidwho-1587490

RESUMEN

Therapeutics that impair the innate immune responses of the liver during the inflammatory cytokine storm like that occurring in COVID-19 are greatly needed. Much interest is currently directed toward Janus kinase (JAK) inhibitors as potential candidates to mitigate this life-threatening complication. Accordingly, this study investigated the influence of the novel JAK inhibitor ruxolitinib (RXB) on concanavalin A (Con A)-induced hepatitis and systemic hyperinflammation in mice to simulate the context occurring in COVID-19 patients. Mice were orally treated with RXB (75 and 150 mg/kg) 2 h prior to the intravenous administration of Con A (20 mg/kg) for a period of 12 h. The results showed that RXB pretreatments were efficient in abrogating Con A-instigated hepatocellular injury (ALT, AST, LDH), necrosis (histopathology), apoptosis (cleaved caspase-3) and nuclear proliferation due to damage (PCNA). The protective mechanism of RXB were attributed to i) prevention of Con A-enhanced hepatic production and systemic release of the proinflammatory cytokines TNF-α, IFN-γ and IL-17A, which coincided with decreasing infiltration of immune cells (monocytes, neutrophils), ii) reducing Con A-induced hepatic overexpression of IL-1ß and CD98 alongside NF-κB activation, and iii) lessening Con A-induced consumption of GSH and GSH peroxidase and generation of oxidative stress products (MDA, 4-HNE, NOx) in the liver. In summary, JAK inhibition by RXB led to eminent protection of the liver against Con A-deleterious manifestations primarily via curbing the inflammatory cytokine storm driven by TNF-α, IFN-γ and IL-17A.


Asunto(s)
Concanavalina A/toxicidad , Síndrome de Liberación de Citoquinas/inducido químicamente , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Nitrilos/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Aldehídos/metabolismo , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas , Relación Dosis-Respuesta a Droga , Inflamación/inducido químicamente , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos BALB C , Nitratos/metabolismo , Nitrilos/administración & dosificación , Nitritos/metabolismo , Estrés Oxidativo , Peroxidasa/metabolismo , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación
6.
Virology ; 566: 60-68, 2022 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1537115

RESUMEN

The emergence of SARS-CoV-2 virus has resulted in a worldwide pandemic, but effective antiviral therapies are not widely available. To improve treatment options, we conducted a high-throughput screen to uncover compounds that block SARS-CoV-2 infection. A minimally pathogenic human betacoronavirus (OC43) was used to infect physiologically-relevant human pulmonary fibroblasts (MRC5) to facilitate rapid antiviral discovery in a preclinical model. Comprehensive profiling was conducted on more than 600 compounds, with each compound arrayed across 10 dose points. Our screening revealed several FDA-approved agents that can attenuate both OC43 and SARS-CoV-2 viral replication, including lapatinib, doramapimod, and 17-AAG. Importantly, lapatinib inhibited SARS-CoV-2 RNA replication by over 50,000-fold. Further, both lapatinib and doramapimod could be combined with remdesivir to improve antiviral activity in cells. These findings reveal novel therapeutic avenues that could limit SARS-CoV-2 infection.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Lapatinib/farmacología , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/farmacología , Alanina/farmacología , Animales , Benzoquinonas/farmacología , COVID-19/virología , Línea Celular , Chlorocebus aethiops , Combinación de Medicamentos , Descubrimiento de Drogas , Sinergismo Farmacológico , Ensayos Analíticos de Alto Rendimiento , Humanos , Lactamas Macrocíclicas/farmacología , Naftalenos/farmacología , Compuestos de Fenilurea/farmacología , Pirazoles/farmacología , ARN Viral/metabolismo , Células Vero , Replicación Viral/efectos de los fármacos
7.
Chem Commun (Camb) ; 57(93): 12476-12479, 2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: covidwho-1500757

RESUMEN

We identified small-molecule enhancers of cellular stress granules by observing molecular crowding of proteins and RNAs in a time-dependent manner. Hit molecules sensitized the IRF3-mediated antiviral mechanism in the presence of poly(I:C) and inhibited the replication of SARS-CoV-2 by inducing stress granule formation. Thus, modulating multimolecular crowding can be a promising strategy against SARS-CoV-2.


Asunto(s)
Antivirales/farmacología , Benzopiranos/farmacología , Gránulos Citoplasmáticos/efectos de los fármacos , Pirazoles/farmacología , SARS-CoV-2/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/química , Benzopiranos/química , Línea Celular Tumoral , Chlorocebus aethiops , Gránulos Citoplasmáticos/metabolismo , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Humanos , Factor 3 Regulador del Interferón/metabolismo , Lopinavir/farmacología , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Poli I-C/farmacología , Pirazoles/química , Relación Estructura-Actividad , Células Vero
8.
Sci Adv ; 7(1)2021 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1388432

RESUMEN

Using AI, we identified baricitinib as having antiviral and anticytokine efficacy. We now show a 71% (95% CI 0.15 to 0.58) mortality benefit in 83 patients with moderate-severe SARS-CoV-2 pneumonia with few drug-induced adverse events, including a large elderly cohort (median age, 81 years). An additional 48 cases with mild-moderate pneumonia recovered uneventfully. Using organotypic 3D cultures of primary human liver cells, we demonstrate that interferon-α2 increases ACE2 expression and SARS-CoV-2 infectivity in parenchymal cells by greater than fivefold. RNA-seq reveals gene response signatures associated with platelet activation, fully inhibited by baricitinib. Using viral load quantifications and superresolution microscopy, we found that baricitinib exerts activity rapidly through the inhibition of host proteins (numb-associated kinases), uniquely among antivirals. This reveals mechanistic actions of a Janus kinase-1/2 inhibitor targeting viral entry, replication, and the cytokine storm and is associated with beneficial outcomes including in severely ill elderly patients, data that incentivize further randomized controlled trials.


Asunto(s)
Antivirales/farmacología , Azetidinas/farmacología , COVID-19/mortalidad , Inhibidores Enzimáticos/farmacología , Quinasas Janus/antagonistas & inhibidores , Hígado/virología , Purinas/farmacología , Pirazoles/farmacología , SARS-CoV-2/patogenicidad , Sulfonamidas/farmacología , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/metabolismo , COVID-19/virología , Síndrome de Liberación de Citoquinas , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Perfilación de la Expresión Génica , Humanos , Interferón alfa-2/metabolismo , Italia , Quinasas Janus/metabolismo , Hígado/efectos de los fármacos , Masculino , Persona de Mediana Edad , Seguridad del Paciente , Activación Plaquetaria , Modelos de Riesgos Proporcionales , RNA-Seq , España , Internalización del Virus/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
9.
Molecules ; 25(19)2020 Oct 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1389458

RESUMEN

A novel series of some hydrazones bearing thiazole moiety were generated via solvent-drop grinding of thiazole carbohydrazide 2 with various carbonyl compounds. Also, dehydrative-cyclocondensation of 2 with active methylene compounds or anhydrides gave the respective pyarzole or pyrazine derivatives. The structures of the newly synthesized compounds were established based on spectroscopic evidences and their alternative syntheses. Additionally, the anti-viral activity of all the products was tested against SARS-CoV-2 main protease (Mpro) using molecular docking combined with molecular dynamics simulation (MDS). The average binding affinities of the compounds 3a, 3b, and 3c (-8.1 ± 0.33 kcal/mol, -8.0 ± 0.35 kcal/mol, and -8.2 ± 0.21 kcal/mol, respectively) are better than that of the positive control Nelfinavir (-6.9 ± 0.51 kcal/mol). This shows the possibility of these three compounds to effectively bind to SARS-CoV-2 Mpro and hence, contradict the virus lifecycle.


Asunto(s)
Antivirales/síntesis química , Betacoronavirus/enzimología , Hidrazonas/síntesis química , Inhibidores de Proteasas/síntesis química , Pirazinas/síntesis química , Pirazoles/síntesis química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Antivirales/farmacología , Betacoronavirus/química , Betacoronavirus/efectos de los fármacos , Sitios de Unión , COVID-19 , Proteasas 3C de Coronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Descubrimiento de Drogas , Humanos , Hidrazonas/farmacología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Pandemias , Neumonía Viral/tratamiento farmacológico , Inhibidores de Proteasas/farmacología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Pirazinas/farmacología , Pirazoles/farmacología , SARS-CoV-2 , Termodinámica , Interfaz Usuario-Computador , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo
11.
Mol Syst Biol ; 17(9): e10426, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1355289

RESUMEN

Although 15-20% of COVID-19 patients experience hyper-inflammation induced by massive cytokine production, cellular triggers of this process and strategies to target them remain poorly understood. Here, we show that the N-terminal domain (NTD) of the SARS-CoV-2 spike protein substantially induces multiple inflammatory molecules in myeloid cells and human PBMCs. Using a combination of phenotypic screening with machine learning-based modeling, we identified and experimentally validated several protein kinases, including JAK1, EPHA7, IRAK1, MAPK12, and MAP3K8, as essential downstream mediators of NTD-induced cytokine production, implicating the role of multiple signaling pathways in cytokine release. Further, we found several FDA-approved drugs, including ponatinib, and cobimetinib as potent inhibitors of the NTD-mediated cytokine release. Treatment with ponatinib outperforms other drugs, including dexamethasone and baricitinib, inhibiting all cytokines in response to the NTD from SARS-CoV-2 and emerging variants. Finally, ponatinib treatment inhibits lipopolysaccharide-mediated cytokine release in myeloid cells in vitro and lung inflammation mouse model. Together, we propose that agents targeting multiple kinases required for SARS-CoV-2-mediated cytokine release, such as ponatinib, may represent an attractive therapeutic option for treating moderate to severe COVID-19.


Asunto(s)
Antivirales/farmacología , Citocinas/metabolismo , Interacciones Huésped-Patógeno/fisiología , Animales , Azetidinas/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Imidazoles/farmacología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Janus Quinasa 1/metabolismo , Lipopolisacáridos/toxicidad , Aprendizaje Automático , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/virología , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Pirazoles/farmacología , Piridazinas/farmacología , Células RAW 264.7 , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo , Sulfonamidas/farmacología
12.
Biomolecules ; 11(7)2021 07 16.
Artículo en Inglés | MEDLINE | ID: covidwho-1323103

RESUMEN

Cyclooxygenase-2 (COX-2) is an important enzyme involved in prostaglandins biosynthesis from arachidonic acid. COX-2 is frequently overexpressed in human cancers and plays a major tumor promoting function. Accordingly, many efforts have been devoted to efficiently target the catalytic site of this enzyme in cancer cells, by using COX-2 specific inhibitors such as celecoxib. However, despite their potent anti-tumor properties, the myriad of detrimental effects associated to the chronic inhibition of COX-2 in healthy tissues, has considerably limited their use in clinic. In addition, increasing evidence indicate that these anti-cancerous properties are not strictly dependent on the inhibition of the catalytic site. These findings have led to the development of non-active COX-2 inhibitors analogues aiming at preserving the antitumor effects of COX-2 inhibitors without their side effects. Among them, two celecoxib derivatives, 2,5-Dimethyl-Celecoxib and OSU-03012, have been developed and suggested for the treatment of viral (e.g., recently SARS-CoV-2), inflammatory, metabolic diseases and cancers. These molecules display stronger anti-tumor properties than celecoxib and thus may represent promising anti-cancer molecules. In this review, we discuss the impact of these two analogues on cancerous processes but also their potential for cancer treatment alone or in combination with existing approaches.


Asunto(s)
Antineoplásicos/uso terapéutico , Celecoxib/uso terapéutico , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Neoplasias/tratamiento farmacológico , Pirazoles/uso terapéutico , Sulfonamidas/uso terapéutico , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/química , Antineoplásicos/farmacología , Celecoxib/efectos adversos , Celecoxib/análogos & derivados , Celecoxib/farmacología , Ciclo Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/efectos adversos , Inhibidores de la Ciclooxigenasa 2/química , Inhibidores de la Ciclooxigenasa 2/farmacología , Humanos , Pirazoles/efectos adversos , Pirazoles/química , Pirazoles/farmacología , Sulfonamidas/efectos adversos , Sulfonamidas/química , Sulfonamidas/farmacología
13.
Sci Rep ; 11(1): 10763, 2021 05 24.
Artículo en Inglés | MEDLINE | ID: covidwho-1242041

RESUMEN

Research on drugs against SARS-CoV-2 (cause of COVID-19) has been one of the major world concerns at present. There have been abundant research data and findings in this field. The interference of drugs on gene expression in cell lines, drug-target, protein-virus receptor networks, and immune cell infiltration of the host may provide useful information for anti-SARS-CoV-2 drug research. To simplify the complex bioinformatics analysis and facilitate the evaluation of the latest research data, we developed OmiczViz ( http://medcode.link/omicsviz ), a web tool that has integrated drug-cell line interference data, virus-host protein-protein interactions, and drug-target interactions. To demonstrate the usages of OmiczViz, we analyzed the gene expression data from cell lines treated with chloroquine and ruxolitinib, the drug-target protein networks of 48 anti-coronavirus drugs and drugs bound with ACE2, and the profiles of immune cell infiltration between different COVID-19 patient groups. Our research shows that chloroquine had a regulatory role of the immune response in renal cell line but not in lung cell line. The anti-coronavirus drug-target network analysis suggested that antihistamine of promethaziney and dietary supplement of Zinc might be beneficial when used jointly with antiviral drugs. The immune infiltration analysis indicated that both the COVID-19 patients admitted to the ICU and the elderly with infection showed immune exhaustion status, yet with different molecular mechanisms. The interactive graphic interface of OmiczViz also makes it easier to analyze newly discovered and user-uploaded data, leading to an in-depth understanding of existing findings and an expansion of existing knowledge of SARS-CoV-2. Collectively, OmicsViz is web program that promotes the research on medical agents against SARS-CoV-2 and supports the evaluation of the latest research findings.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Interfaz Usuario-Computador , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , COVID-19/metabolismo , COVID-19/patología , COVID-19/virología , Línea Celular , Cloroquina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Internet , Nitrilos , Unión Proteica , Pirazoles/farmacología , Pirimidinas , Índice de Severidad de la Enfermedad
14.
J Hepatol ; 75(3): 647-658, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1228069

RESUMEN

BACKGROUND AND AIMS: COVID-19 is associated with liver injury and elevated interleukin-6 (IL-6). We hypothesized that IL-6 trans-signaling in liver sinusoidal endothelial cells (LSECs) leads to endotheliopathy (a proinflammatory and procoagulant state) and liver injury in COVID-19. METHODS: Coagulopathy, endotheliopathy, and alanine aminotransferase (ALT) were retrospectively analyzed in a subset (n = 68), followed by a larger cohort (n = 3,780) of patients with COVID-19. Liver histology from 43 patients with COVID-19 was analyzed for endotheliopathy and its relationship to liver injury. Primary human LSECs were used to establish the IL-6 trans-signaling mechanism. RESULTS: Factor VIII, fibrinogen, D-dimer, von Willebrand factor (vWF) activity/antigen (biomarkers of coagulopathy/endotheliopathy) were significantly elevated in patients with COVID-19 and liver injury (elevated ALT). IL-6 positively correlated with vWF antigen (p = 0.02), factor VIII activity (p = 0.02), and D-dimer (p <0.0001). On liver histology, patients with COVID-19 and elevated ALT had significantly increased vWF and platelet staining, supporting a link between liver injury, coagulopathy, and endotheliopathy. Intralobular neutrophils positively correlated with platelet (p <0.0001) and vWF (p <0.01) staining, and IL-6 levels positively correlated with vWF staining (p <0.01). IL-6 trans-signaling leads to increased expression of procoagulant (factor VIII, vWF) and proinflammatory factors, increased cell surface vWF (p <0.01), and increased platelet attachment in LSECs. These effects were blocked by soluble glycoprotein 130 (IL-6 trans-signaling inhibitor), the JAK inhibitor ruxolitinib, and STAT1/3 small-interfering RNA knockdown. Hepatocyte fibrinogen expression was increased by the supernatant of LSECs subjected to IL-6 trans-signaling. CONCLUSION: IL-6 trans-signaling drives the coagulopathy and hepatic endotheliopathy associated with COVID-19 and could be a possible mechanism behind liver injury in these patients. LAY SUMMARY: Patients with SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infection often have liver injury, but why this occurs remains unknown. High levels of interleukin-6 (IL-6) and its circulating receptor, which form a complex to induce inflammatory signals, have been observed in patients with COVID-19. This paper demonstrates that the IL-6 signaling complex causes harmful changes to liver sinusoidal endothelial cells and may promote blood clotting and contribute to liver injury.


Asunto(s)
COVID-19/complicaciones , Células Endoteliales/patología , Interleucina-6/fisiología , Hepatopatías/etiología , SARS-CoV-2 , Adulto , Trastornos de la Coagulación Sanguínea/etiología , Fibrinógeno/análisis , Humanos , Interleucina-6/sangre , Janus Quinasa 1/metabolismo , Nitrilos , Pirazoles/farmacología , Pirimidinas , Estudios Retrospectivos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Factor de von Willebrand/análisis
15.
J Clin Pharmacol ; 61(10): 1274-1285, 2021 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1192122

RESUMEN

Baricitinib is a JAK1/2 inhibitor that was first approved for treating moderate to severe rheumatoid arthritis (RA) but that later showed considerable efficacy in the control of exaggerated inflammatory responses that occur in a wide range of diseases. There is a growing body of evidence, obtained from clinical trials and case reports, demonstrating clinical and paraclinical improvement in patients following administration of baricitinib including RA, systemic lupus erythematosus, psoriasis, atopic dermatitis, alopecia areata, interferon-mediated autoinflammatory diseases, graft-versus-host disease, diabetic kidney disease, and, recently, coronavirus disease-19. However, despite overall encouraging results, many adverse effects have been observed in baricitinib-treated patients, ranging from simple infections to increased risk of malignancies, particularly in long-term use. The significant efficacy of baricitinib, versus the probable adverse effects, urge further investigation before establishing it as a part of standard therapeutic protocols. Here, we have provided a review of the studies that have used baricitinib for treating various inflammatory disorders and summarized the advantages and disadvantages of its administration.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Azetidinas/farmacología , Tratamiento Farmacológico de COVID-19 , COVID-19 , Inflamación/tratamiento farmacológico , Purinas/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología , COVID-19/inmunología , Humanos , Inhibidores de las Cinasas Janus/farmacología , Medición de Riesgo , SARS-CoV-2 , Resultado del Tratamiento
16.
Mol Ther ; 29(3): 1174-1185, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: covidwho-985497

RESUMEN

Self-amplifying RNA (saRNA) is a cutting-edge platform for both nucleic acid vaccines and therapeutics. saRNA is self-adjuvanting, as it activates types I and III interferon (IFN), which enhances the immunogenicity of RNA vaccines but can also lead to inhibition of translation. In this study, we screened a library of saRNA constructs with cis-encoded innate inhibiting proteins (IIPs) and determined the effect on protein expression and immunogenicity. We observed that the PIV-5 V and Middle East respiratory syndrome coronavirus (MERS-CoV) ORF4a proteins enhance protein expression 100- to 500-fold in vitro in IFN-competent HeLa and MRC5 cells. We found that the MERS-CoV ORF4a protein partially abates dose nonlinearity in vivo, and that ruxolitinib, a potent Janus kinase (JAK)/signal transducer and activator of transcription (STAT) inhibitor, but not the IIPs, enhances protein expression of saRNA in vivo. Both the PIV-5 V and MERS-CoV ORF4a proteins were found to enhance the percentage of resident cells in human skin explants expressing saRNA and completely rescued dose nonlinearity of saRNA. Finally, we observed that the MERS-CoV ORF4a increased the rabies virus (RABV)-specific immunoglobulin G (IgG) titer and neutralization half-maximal inhibitory concentration (IC50) by ∼10-fold in rabbits, but not in mice or rats. These experiments provide a proof of concept that IIPs can be directly encoded into saRNA vectors and effectively abate the nonlinear dose dependency and enhance immunogenicity.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , Inmunogenicidad Vacunal , Biosíntesis de Proteínas/efectos de los fármacos , Vacunas Sintéticas/farmacología , Proteínas del Envoltorio Viral/administración & dosificación , Animales , Línea Celular , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Virus de la Encefalitis Equina Venezolana/inmunología , Virus de la Encefalitis Equina Venezolana/patogenicidad , Fibroblastos , Regulación de la Expresión Génica , Células HeLa , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunoglobulina G/biosíntesis , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/genética , Quinasas Janus/inmunología , Ratones , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/patogenicidad , FN-kappa B/genética , FN-kappa B/inmunología , Nitrilos , Virus de la Parainfluenza 5/efectos de los fármacos , Virus de la Parainfluenza 5/inmunología , Virus de la Parainfluenza 5/patogenicidad , Pirazoles/farmacología , Pirimidinas , Conejos , Virus de la Rabia/efectos de los fármacos , Virus de la Rabia/inmunología , Virus de la Rabia/patogenicidad , Ratas , Factores de Transcripción STAT/antagonistas & inhibidores , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/inmunología , Transducción de Señal , Vacunas Sintéticas/biosíntesis , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
17.
Sci Rep ; 11(1): 2512, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: covidwho-1054059

RESUMEN

Whenever some phenomenon can be represented as a graph or a network it seems pertinent to explore how much the mathematical properties of that network impact the phenomenon. In this study we explore the same philosophy in the context of immunology. Our objective was to assess the correlation of "size" (number of edges and minimum vertex cover) of the JAK/STAT network with treatment effect in rheumatoid arthritis (RA), phenotype of viral infection and effect of immunosuppressive agents on a system infected with the coronavirus. We extracted the JAK/STAT pathway from Kyoto Encyclopedia of Genes and Genomes (KEGG, hsa04630). The effects of the following drugs, and their combinations, commonly used in RA were tested: methotrexate, prednisolone, rituximab, tocilizumab, tofacitinib and baricitinib. Following viral systems were also tested for their ability to evade the JAK/STAT pathway: Measles, Influenza A, West Nile virus, Japanese B virus, Yellow Fever virus, respiratory syncytial virus, Kaposi's sarcoma virus, Hepatitis B and C virus, cytomegalovirus, Hendra and Nipah virus and Coronavirus. Good correlation of edges and minimum vertex cover with clinical efficacy were observed (for edge, rho = - 0.815, R2 = 0.676, p = 0.007, for vertex cover rho = - 0.793, R2 = 0.635, p = 0.011). In the viral systems both edges and vertex cover were associated with acuteness of viral infections. In the JAK/STAT system already infected with coronavirus, maximum reduction in size was achieved with baricitinib. To conclude, algebraic and combinatorial invariant of a network may explain its biological behaviour. At least theoretically, baricitinib may be an attractive target for treatment of coronavirus infection.


Asunto(s)
Artritis Reumatoide/metabolismo , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Virosis/tratamiento farmacológico , Virosis/metabolismo , Anticuerpos Monoclonales Humanizados/farmacología , Artritis Reumatoide/genética , Azetidinas/farmacología , Redes Reguladoras de Genes , Humanos , Quinasas Janus/genética , Metotrexato/farmacología , Modelos Estadísticos , Piperidinas/farmacología , Prednisolona/farmacología , Purinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Rituximab/farmacología , Factores de Transcripción STAT/genética , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
19.
Ann Palliat Med ; 10(1): 707-720, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: covidwho-1030457

RESUMEN

The whole world is battling through coronavirus disease 2019 (COVID-19) which is a fatal pandemic. In the early 2020, the World Health Organization (WHO) declared it as a global health emergency without definitive treatments and preventive approaches. In the absence of definitive therapeutic agents, this thorough review summarizes and outlines the potency and safety of all molecules and therapeutics which may have potential antiviral effects. A number of molecules and therapeutics licensed or being tested for some other conditions were found effective in different in vitro studies as well as in many small sample-sized clinical trials and independent case studies. However, in those clinical trials, there were some limitations which need to be overcome to find the most promising antiviral against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In conclusion, many of above-mentioned antivirals seems to have some therapeutic effects but none of them have been shown to have a strong evidence for their proper recommendation and approval in the treatment of COVID-19. Constantly evolving new evidences, exclusive adult data, language barrier, and type of study (observational, retrospective, small-sized clinical trials, or independent case series) resulted to the several limitations of this review. The need for multicentered, large sample-sized, randomized, placebo-controlled trials on COVID-19 patients to reach a proper conclusion on the most promising antiviral agent is warranted.


Asunto(s)
Antivirales/uso terapéutico , COVID-19/terapia , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/análogos & derivados , Alanina/farmacología , Alanina/uso terapéutico , Amidas/farmacología , Amidas/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Azetidinas/farmacología , Azetidinas/uso terapéutico , Cloroquina/farmacología , Cloroquina/uso terapéutico , Combinación de Medicamentos , Humanos , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Inmunización Pasiva , Indoles/farmacología , Indoles/uso terapéutico , Interferones/farmacología , Interferones/uso terapéutico , Ivermectina/farmacología , Ivermectina/uso terapéutico , Lopinavir/farmacología , Lopinavir/uso terapéutico , Nitrocompuestos , Oseltamivir/farmacología , Oseltamivir/uso terapéutico , Purinas/farmacología , Purinas/uso terapéutico , Pirazinas/farmacología , Pirazinas/uso terapéutico , Pirazoles/farmacología , Pirazoles/uso terapéutico , Ribavirina/farmacología , Ribavirina/uso terapéutico , Ritonavir/farmacología , Ritonavir/uso terapéutico , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Tiazoles/farmacología , Tiazoles/uso terapéutico , Sueroterapia para COVID-19
20.
Infect Genet Evol ; 85: 104419, 2020 11.
Artículo en Inglés | MEDLINE | ID: covidwho-997272

RESUMEN

The COVID-19 pandemic, caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is a current global threat for which there is an urgent need to search for an effective therapy. The transmembrane spike (S) glycoprotein of SARS-CoV-2 directly binds to the host angiotensin-converting enzyme 2 (ACE2) and mediates viral entrance, which is therefore considered as a promising drug target. Considering that new drug development is a time-consuming process, drug repositioning may facilitate rapid drug discovery dealing with sudden infectious diseases. Here, we compared the differences between the virtual structural proteins of SARS-CoV-2 and SARS-CoV, and selected a pocket mainly localizing in the fusion cores of S2 domain for drug screening. A virtual drug design algorithm screened the Food and Drug Administration-approved drug library of 1234 compounds, and 13 top scored compounds were obtained through manual screening. Through in vitro molecular interaction experiments, eltrombopag was further verified to possess a high binding affinity to S protein plus human ACE2 and could potentially affect the stability of the ACE2-S protein complex. Hence, it is worth further exploring eltrombopag as a potential drug for the treatment of SARS-CoV-2 infection.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Benzoatos/farmacología , Hidrazinas/farmacología , Pirazoles/farmacología , SARS-CoV-2/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Algoritmos , Enzima Convertidora de Angiotensina 2/química , Benzoatos/química , Simulación por Computador , Diseño de Fármacos , Reposicionamiento de Medicamentos , Humanos , Hidrazinas/química , Modelos Moleculares , Unión Proteica , Estabilidad Proteica , Pirazoles/química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA