Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Int J Mol Sci ; 23(6)2022 Mar 16.
Artículo en Inglés | MEDLINE | ID: covidwho-1742493

RESUMEN

Advanced prostate cancer (PCa) patients with bone metastases are treated with androgen pathway directed therapy (APDT). However, this treatment invariably fails and the cancer becomes castration resistant. To elucidate resistance mechanisms and to provide a more predictive pre-clinical research platform reflecting tumor heterogeneity, we established organoids from a patient-derived xenograft (PDX) model of bone metastatic prostate cancer, PCSD1. APDT-resistant PDX-derived organoids (PDOs) emerged when cultured without androgen or with the anti-androgen, enzalutamide. Transcriptomics revealed up-regulation of neurogenic and steroidogenic genes and down-regulation of DNA repair, cell cycle, circadian pathways and the severe acute respiratory syndrome (SARS)-CoV-2 host viral entry factors, ACE2 and TMPRSS2. Time course analysis of the cell cycle in live cells revealed that enzalutamide induced a gradual transition into a reversible dormant state as shown here for the first time at the single cell level in the context of multi-cellular, 3D living organoids using the Fucci2BL fluorescent live cell cycle tracker system. We show here a new mechanism of castration resistance in which enzalutamide induced dormancy and novel basal-luminal-like cells in bone metastatic prostate cancer organoids. These PDX organoids can be used to develop therapies targeting dormant APDT-resistant cells and host factors required for SARS-CoV-2 viral entry.


Asunto(s)
Neoplasias Óseas/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Organoides/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/genética , Andrógenos/farmacología , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Benzamidas/farmacología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , COVID-19/genética , COVID-19/metabolismo , COVID-19/virología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Nitrilos/farmacología , Feniltiohidantoína/farmacología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Virales/genética , Receptores Virales/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Trasplante Heterólogo , Internalización del Virus
2.
JCI Insight ; 7(2)2022 01 25.
Artículo en Inglés | MEDLINE | ID: covidwho-1649609

RESUMEN

Cellular and molecular mechanisms driving morbidity following SARS-CoV-2 infection have not been well defined. The receptor for advanced glycation end products (RAGE) is a central mediator of tissue injury and contributes to SARS-CoV-2 disease pathogenesis. In this study, we temporally delineated key cell and molecular events leading to lung injury in mice following SARS-CoV-2 infection and assessed efficacy of therapeutically targeting RAGE to improve survival. Early following infection, SARS-CoV-2 replicated to high titers within the lungs and evaded triggering inflammation and cell death. However, a significant necrotic cell death event in CD45- populations, corresponding with peak viral loads, was observed on day 2 after infection. Metabolic reprogramming and inflammation were initiated following this cell death event and corresponded with increased lung interstitial pneumonia, perivascular inflammation, and endothelial hyperplasia together with decreased oxygen saturation. Therapeutic treatment with the RAGE antagonist FPS-ZM1 improved survival in infected mice and limited inflammation and associated perivascular pathology. Together, these results provide critical characterization of disease pathogenesis in the mouse model and implicate a role for RAGE signaling as a therapeutic target to improve outcomes following SARS-CoV-2 infection.


Asunto(s)
Benzamidas/farmacología , Tratamiento Farmacológico de COVID-19 , COVID-19 , Pulmón , Receptor para Productos Finales de Glicación Avanzada , SARS-CoV-2/fisiología , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , COVID-19/genética , COVID-19/metabolismo , Modelos Animales de Enfermedad , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Transgénicos , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo
3.
J Med Chem ; 65(1): 876-884, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: covidwho-1606194

RESUMEN

Coronavirus disease 2019 (COVID-19) pandemic, a global health threat, was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The SARS-CoV-2 papain-like cysteine protease (PLpro) was recognized as a promising drug target because of multiple functions in virus maturation and antiviral immune responses. Inhibitor GRL0617 occupied the interferon-stimulated gene 15 (ISG15) C-terminus-binding pocket and showed an effective antiviral inhibition. Here, we described a novel peptide-drug conjugate (PDC), in which GRL0617 was linked to a sulfonium-tethered peptide derived from PLpro-specific substrate LRGG. The EM-C and EC-M PDCs showed a promising in vitro IC50 of 7.40 ± 0.37 and 8.63 ± 0.55 µM, respectively. EC-M could covalently label PLpro active site C111 and display anti-ISGylation activities in cellular assays. The results represent the first attempt to design PDCs composed of stabilized peptide inhibitors and GRL0617 to inhibit PLpro. These novel PDCs provide promising opportunities for antiviral drug design.


Asunto(s)
Compuestos de Anilina/química , Antivirales/metabolismo , Benzamidas/química , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Diseño de Fármacos , Naftalenos/química , Péptidos/química , SARS-CoV-2/enzimología , Compuestos de Anilina/metabolismo , Compuestos de Anilina/farmacología , Antivirales/química , Antivirales/farmacología , Antivirales/uso terapéutico , Benzamidas/metabolismo , Benzamidas/farmacología , COVID-19/patología , COVID-19/virología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Proteasas Similares a la Papaína de Coronavirus/química , Citocinas/química , Evaluación Preclínica de Medicamentos , Humanos , Concentración 50 Inhibidora , Naftalenos/metabolismo , Naftalenos/farmacología , SARS-CoV-2/aislamiento & purificación , Ubiquitinas/química , Tratamiento Farmacológico de COVID-19
4.
Int J Mol Sci ; 22(24)2021 Dec 18.
Artículo en Inglés | MEDLINE | ID: covidwho-1580690

RESUMEN

Since the start of the COVID-19 outbreak, pharmaceutical companies and research groups have focused on the development of vaccines and antiviral drugs against SARS-CoV-2. Here, we apply a drug repurposing strategy to identify drug candidates that are able to block the entrance of the virus into human cells. By combining virtual screening with in vitro pseudovirus assays and antiviral assays in Human Lung Tissue (HLT) cells, we identify entrectinib as a potential antiviral drug.


Asunto(s)
Benzamidas/farmacología , Tratamiento Farmacológico de COVID-19 , Indazoles/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/farmacología , Benzamidas/metabolismo , COVID-19/metabolismo , Línea Celular , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos/métodos , Humanos , Indazoles/metabolismo , Pulmón/patología , Pulmón/virología , Simulación del Acoplamiento Molecular , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Células Vero , Acoplamiento Viral/efectos de los fármacos
5.
Neuropharmacology ; 207: 108935, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1586929

RESUMEN

AIMS: Acetaminophen or paracetamol (PAR), the recommended antipyretic in COVID-19 and clinically used to alleviate stroke-associated hyperthermia interestingly activates cannabinoid receptor (CB1) through its AM404 metabolite, however, to date, no study reports the in vivo activation of PAR/AM404/CB1 axis in stroke. The current study deciphers the neuroprotective effect off PAR in cerebral ischemia/reperfusion (IR) rat model and unmasks its link with AM404/CB1/PI3K/Akt axis. MATERIALS AND METHODS: Animals were allocated into 5 groups: (I) sham-operated (SO), (II) IR, (III) IR + PAR (100 mg/kg), (IV) IR + PAR (100 mg/kg) + URB597; anandamide degradation inhibitor (0.3 mg/kg) and (V) IR + PAR (100 mg/kg) + AM4113; CB1 Blocker (5 mg/kg). All drugs were intraperitoneally administered at the inception of the reperfusion period. KEY FINDINGS: PAR administration alleviated the cognitive impairment in the Morris Water Maze as well as hippocampal histopathological and immunohistochemical examination of GFAP. The PAR signaling was associated with elevation of anandamide level, CB1 receptor expression and survival proteins as pS473-Akt. P(tyr202/thr204)-ERK1/2 and pS9-GSK3ß. Simultaneously, PAR increased hippocampal BDNF and ß-arrestin1 levels and decreased glutamate level. PAR restores the deranged redox milieu induced by IR Injury, by reducing lipid peroxides, myeloperoxidase activity and NF-κB and increasing NPSH, total antioxidant capacity, nitric oxide and Nrf2 levels. The pre-administration of AM4113 reversed PAR effects, while URB597 potentiated them. SIGNIFICANCE: PAR poses a significant neuroprotective effect which may be mediated, at least in part, via activation of anandamide/CB1/PI3K/Akt pathway in the IR rat model.


Asunto(s)
Acetaminofén/farmacología , Antipiréticos/farmacología , Benzamidas/farmacología , Carbamatos/farmacología , Inhibidores Enzimáticos/farmacología , Hipocampo/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Receptor Cannabinoide CB1/metabolismo , Daño por Reperfusión/metabolismo , Amidohidrolasas/antagonistas & inhibidores , Animales , Ácidos Araquidónicos/metabolismo , Antagonistas de Receptores de Cannabinoides/farmacología , Endocannabinoides/metabolismo , Hipocampo/irrigación sanguínea , Hipocampo/metabolismo , Hipocampo/fisiopatología , Fosfatidilinositol 3-Quinasas/metabolismo , Alcamidas Poliinsaturadas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Daño por Reperfusión/fisiopatología
6.
Sci Rep ; 11(1): 23315, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1550334

RESUMEN

The COVID-19 pandemic has highlighted the urgent need for the identification of new antiviral drug therapies for a variety of diseases. COVID-19 is caused by infection with the human coronavirus SARS-CoV-2, while other related human coronaviruses cause diseases ranging from severe respiratory infections to the common cold. We developed a computational approach to identify new antiviral drug targets and repurpose clinically-relevant drug compounds for the treatment of a range of human coronavirus diseases. Our approach is based on graph convolutional networks (GCN) and involves multiscale host-virus interactome analysis coupled to off-target drug predictions. Cell-based experimental assessment reveals several clinically-relevant drug repurposing candidates predicted by the in silico analyses to have antiviral activity against human coronavirus infection. In particular, we identify the MET inhibitor capmatinib as having potent and broad antiviral activity against several coronaviruses in a MET-independent manner, as well as novel roles for host cell proteins such as IRAK1/4 in supporting human coronavirus infection, which can inform further drug discovery studies.


Asunto(s)
Antivirales/farmacología , Coronavirus/efectos de los fármacos , Coronavirus/metabolismo , Desarrollo de Medicamentos/métodos , Reposicionamiento de Medicamentos/métodos , Benzamidas/farmacología , Línea Celular , Simulación por Computador , Coronavirus/química , Bases de Datos Farmacéuticas , Descubrimiento de Drogas/métodos , Interacciones Huésped-Patógeno , Humanos , Imidazoles/farmacología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , SARS-CoV-2/química , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Triazinas/farmacología , Tratamiento Farmacológico de COVID-19
7.
EBioMedicine ; 74: 103712, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: covidwho-1536515

RESUMEN

BACKGROUND: Despite clinical success with anti-spike vaccines, the effectiveness of neutralizing antibodies and vaccines has been compromised by rapidly spreading SARS-CoV-2 variants. Viruses can hijack the glycosylation machinery of host cells to shield themselves from the host's immune response and attenuate antibody efficiency. However, it remains unclear if targeting glycosylation on viral spike protein can impair infectivity of SARS-CoV-2 and its variants. METHODS: We adopted flow cytometry, ELISA, and BioLayer interferometry approaches to assess binding of glycosylated or deglycosylated spike with ACE2. Viral entry was determined by luciferase, immunoblotting, and immunofluorescence assays. Genome-wide association study (GWAS) revealed a significant relationship between STT3A and COVID-19 severity. NF-κB/STT3A-regulated N-glycosylation was investigated by gene knockdown, chromatin immunoprecipitation, and promoter assay. We developed an antibody-drug conjugate (ADC) that couples non-neutralization anti-spike antibody with NGI-1 (4G10-ADC) to specifically target SARS-CoV-2-infected cells. FINDINGS: The receptor binding domain and three distinct SARS-CoV-2 surface N-glycosylation sites among 57,311 spike proteins retrieved from the NCBI-Virus-database are highly evolutionarily conserved (99.67%) and are involved in ACE2 interaction. STT3A is a key glycosyltransferase catalyzing spike glycosylation and is positively correlated with COVID-19 severity. We found that inhibiting STT3A using N-linked glycosylation inhibitor-1 (NGI-1) impaired SARS-CoV-2 infectivity and that of its variants [Alpha (B.1.1.7) and Beta (B.1.351)]. Most importantly, 4G10-ADC enters SARS-CoV-2-infected cells and NGI-1 is subsequently released to deglycosylate spike protein, thereby reinforcing the neutralizing abilities of antibodies, vaccines, or convalescent sera and reducing SARS-CoV-2 variant infectivity. INTERPRETATION: Our results indicate that targeting evolutionarily-conserved STT3A-mediated glycosylation via an ADC can exert profound impacts on SARS-CoV-2 variant infectivity. Thus, we have identified a novel deglycosylation method suitable for eradicating SARS-CoV-2 variant infection in vitro. FUNDING: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.


Asunto(s)
Benzamidas/farmacología , Tratamiento Farmacológico de COVID-19 , Glicosilación/efectos de los fármacos , Hexosiltransferasas/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Sulfonamidas/farmacología , Internalización del Virus/efectos de los fármacos , Células A549 , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Línea Celular , Células HEK293 , Hexosiltransferasas/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , SARS-CoV-2/crecimiento & desarrollo , Glicoproteína de la Espiga del Coronavirus/metabolismo
8.
Mol Divers ; 26(4): 2189-2209, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: covidwho-1442141

RESUMEN

Papain-like protease (nsp-3; non-structural protein) of novel corona virus is an ideal target for developing drugs as it plays multiple important functions for viral growth and replication. For instance, role of nsp-3 has been recognized in cleavage of viral polyprotein; furthermore, in infected host it weakens the immune system via downregulating the production of type I interferon. This downregulation is promoted by removal of ubiquitin-like interferon-stimulated gene 15 protein (ISG15) from interferon-responsive factor 3 (IRF3) protein. Among known inhibitors of SARS-CoV-PLpro GRL0617 is by far the most effective inhibitor. As PLpro of SARS-CoV2 is having more than 80% similarity with SARS-CoV-PLpro, GRL0617 is reported to be effective even against SARS-CoV2. Owing to this similarity, certain key amino acids remain the same/conserved in both proteins. Among conserved amino acids Tyr268 for SARS-CoV2 and Tyr269 for SARS-CoV produce important hydrophobic interactions with aromatic rings of GRL0617. Here, in this study antibacterial compounds were collected from ZINC database, and they were filtered to select compounds that are having similar structural features as GRL0617. This filtered library of compound was then docked with SARS-CoV and CoV2-PLpro. Five hits were noted that were able to interact with Tyr268 (SARS-CoV2) and Tyr269 (SARS-CoV). Further, best hit 2-(2-((benzofuran-2-carboxamido)methyl)-5-methoxy-1H-indol-1-yl)acetic acid (ZINC44459905) was studied using molecular dynamic simulation where stability of protein-ligand complex as well as stability of produced interactions was noted.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Proteasas Similares a la Papaína de Coronavirus , Reposicionamiento de Medicamentos , SARS-CoV-2 , Aminoácidos , Compuestos de Anilina/farmacología , Antibacterianos , Benzamidas/farmacología , Proteasas Similares a la Papaína de Coronavirus/antagonistas & inhibidores , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Naftalenos/farmacología , ARN Viral , SARS-CoV-2/efectos de los fármacos , Ubiquitinas/química , Ubiquitinas/metabolismo
9.
Nat Commun ; 12(1): 4068, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1294463

RESUMEN

SARS-CoV-2 attacks various organs, most destructively the lung, and cellular entry requires two host cell surface proteins: ACE2 and TMPRSS2. Downregulation of one or both of these is thus a potential therapeutic approach for COVID-19. TMPRSS2 is a known target of the androgen receptor, a ligand-activated transcription factor; androgen receptor activation increases TMPRSS2 levels in various tissues, most notably prostate. We show here that treatment with the antiandrogen enzalutamide-a well-tolerated drug widely used in advanced prostate cancer-reduces TMPRSS2 levels in human lung cells and in mouse lung. Importantly, antiandrogens significantly reduced SARS-CoV-2 entry and infection in lung cells. In support of this experimental data, analysis of existing datasets shows striking co-expression of AR and TMPRSS2, including in specific lung cell types targeted by SARS-CoV-2. Together, the data presented provides strong evidence to support clinical trials to assess the efficacy of antiandrogens as a treatment option for COVID-19.


Asunto(s)
Antagonistas de Andrógenos/farmacología , Benzamidas/farmacología , Tratamiento Farmacológico de COVID-19 , Nitrilos/farmacología , Feniltiohidantoína/farmacología , Serina Endopeptidasas/metabolismo , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/síntesis química , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/metabolismo , COVID-19/virología , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Pulmón/metabolismo , Pulmón/virología , Masculino , Ratones , SARS-CoV-2/efectos de los fármacos , Serina Endopeptidasas/genética
10.
Biochem J ; 478(13): 2517-2531, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: covidwho-1290988

RESUMEN

The COVID-19 pandemic has emerged as the biggest life-threatening disease of this century. Whilst vaccination should provide a long-term solution, this is pitted against the constant threat of mutations in the virus rendering the current vaccines less effective. Consequently, small molecule antiviral agents would be extremely useful to complement the vaccination program. The causative agent of COVID-19 is a novel coronavirus, SARS-CoV-2, which encodes at least nine enzymatic activities that all have drug targeting potential. The papain-like protease (PLpro) contained in the nsp3 protein generates viral non-structural proteins from a polyprotein precursor, and cleaves ubiquitin and ISG protein conjugates. Here we describe the expression and purification of PLpro. We developed a protease assay that was used to screen a custom compound library from which we identified dihydrotanshinone I and Ro 08-2750 as compounds that inhibit PLpro in protease and isopeptidase assays and also inhibit viral replication in cell culture-based assays.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Proteasas Similares a la Papaína de Coronavirus/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos , SARS-CoV-2/enzimología , Bibliotecas de Moléculas Pequeñas/farmacología , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Compuestos de Anilina/farmacología , Animales , Benzamidas/farmacología , Chlorocebus aethiops , Proteasas Similares a la Papaína de Coronavirus/genética , Proteasas Similares a la Papaína de Coronavirus/aislamiento & purificación , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Sinergismo Farmacológico , Pruebas de Enzimas , Flavinas/farmacología , Transferencia Resonante de Energía de Fluorescencia , Furanos/farmacología , Ensayos Analíticos de Alto Rendimiento , Concentración 50 Inhibidora , Naftalenos/farmacología , Fenantrenos/farmacología , Quinonas/farmacología , Reproducibilidad de los Resultados , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/crecimiento & desarrollo , Bibliotecas de Moléculas Pequeñas/química , Células Vero , Replicación Viral/efectos de los fármacos
11.
Sci Rep ; 11(1): 11130, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: covidwho-1246392

RESUMEN

The sex discordance in COVID-19 outcomes has been widely recognized, with males generally faring worse than females and a potential link to sex steroids. A plausible mechanism is androgen-induced expression of TMPRSS2 and/or ACE2 in pulmonary tissues that may increase susceptibility or severity in males. This hypothesis is the subject of several clinical trials of anti-androgen therapies around the world. Here, we investigated the sex-associated TMPRSS2 and ACE2 expression in human and mouse lungs and interrogated the possibility of pharmacologic modification of their expression with anti-androgens. We found no evidence for increased TMPRSS2 expression in the lungs of males compared to females in humans or mice. Furthermore, in male mice, treatment with the androgen receptor antagonist enzalutamide did not decrease pulmonary TMPRSS2. On the other hand, ACE2 and AR expression was sexually dimorphic and higher in males than females. ACE2 was moderately suppressible with enzalutamide administration. Our work suggests that sex differences in COVID-19 outcomes attributable to viral entry are independent of TMPRSS2. Modest changes in ACE2 could account for some of the sex discordance.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , Pulmón/efectos de los fármacos , Receptores Androgénicos/metabolismo , Serina Endopeptidasas/metabolismo , Antagonistas de Receptores Androgénicos/farmacología , Andrógenos , Enzima Convertidora de Angiotensina 2/genética , Animales , Benzamidas/farmacología , COVID-19/genética , Línea Celular Tumoral , Secuenciación de Inmunoprecipitación de Cromatina , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Inmunohistoquímica , Pulmón/metabolismo , Pulmón/virología , Masculino , Ratones , Nitrilos/farmacología , Feniltiohidantoína/farmacología , Serina Endopeptidasas/genética , Fumadores
12.
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
13.
mSphere ; 6(1)2021 02 03.
Artículo en Inglés | MEDLINE | ID: covidwho-1063056

RESUMEN

Smallpox, caused by Variola virus (VARV), was eradicated in 1980; however, VARV bioterrorist threats still exist, necessitating readily available therapeutics. Current preparedness activities recognize the importance of oral antivirals and recommend therapeutics with different mechanisms of action. Monkeypox virus (MPXV) is closely related to VARV, causing a highly similar clinical human disease, and can be used as a surrogate for smallpox antiviral testing. The prairie dog MPXV model has been characterized and used to study the efficacy of antipoxvirus therapeutics, including recently approved TPOXX (tecovirimat). Brincidofovir (BCV; CMX001) has shown antiviral activity against double-stranded DNA viruses, including poxviruses. To determine the exposure of BCV following oral administration to prairie dogs, a pharmacokinetics (PK) study was performed. Analysis of BCV plasma concentrations indicated variability, conceivably due to the outbred nature of the animals. To determine BCV efficacy in the MPXV prairie dog model, groups of animals were intranasally challenged with 9 × 105 plaque-forming units (PFU; 90% lethal dose [LD90]) of MPXV on inoculation day 0 (ID0). Animals were divided into groups based on the first day of BCV treatment relative to inoculation day (ID-1, ID0, or ID1). A trend in efficacy was noted dependent upon treatment initiation (57% on ID-1, 43% on ID0, and 29% on ID1) but was lower than demonstrated in other animal models. Analysis of the PK data indicated that BCV plasma exposure (maximum concentration [Cmax]) and the time of the last quantifiable concentration (AUClast) were lower than in other animal models administered the same doses, indicating that suboptimal BCV exposure may explain the lower protective effect on survival.IMPORTANCE Preparedness activities against highly transmissible viruses with high mortality rates have been highlighted during the ongoing coronavirus disease 2019 (COVID-19) pandemic. Smallpox, caused by variola virus (VARV) infection, is highly transmissible, with an estimated 30% mortality. Through an intensive vaccination campaign, smallpox was declared eradicated in 1980, and routine smallpox vaccination of individuals ceased. Today's current population has little/no immunity against VARV. If smallpox were to reemerge, the worldwide results would be devastating. Recent FDA approval of one smallpox antiviral (tecovirimat) was a successful step in biothreat preparedness; however, orthopoxviruses can become resistant to treatment, suggesting the need for multiple therapeutics. Our paper details the efficacy of the investigational smallpox drug brincidofovir in a monkeypox virus (MPXV) animal model. Since brincidofovir has not been tested in vivo against smallpox, studies with the related virus MPXV are critical in understanding whether it would be protective in the event of a smallpox outbreak.


Asunto(s)
Citosina/análogos & derivados , Monkeypox virus/efectos de los fármacos , Organofosfonatos/farmacología , Organofosfonatos/farmacocinética , Viruela/tratamiento farmacológico , Animales , Antivirales/farmacocinética , Antivirales/farmacología , Benzamidas/farmacocinética , Benzamidas/farmacología , Citosina/farmacocinética , Citosina/farmacología , Modelos Animales de Enfermedad , Perros , Femenino , Isoindoles/farmacocinética , Isoindoles/farmacología , Masculino , Virus de la Viruela/efectos de los fármacos
14.
Mol Cancer Res ; 19(4): 549-554, 2021 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1058113

RESUMEN

The outbreak of the novel coronavirus disease 2019 (COVID-19) has emerged as one of the biggest global health threats worldwide. As of October 2020, more than 44 million confirmed cases and more than 1,160,000 deaths have been reported globally, and the toll is likely to be much higher before the pandemic is over. There are currently little therapeutic options available and new potential targets are intensively investigated. Recently, Bruton tyrosine kinase (BTK) has emerged as an interesting candidate. Elevated levels of BTK activity have been reported in blood monocytes from patients with severe COVID-19, compared with those from healthy volunteers. Importantly, various studies confirmed empirically that administration of BTK inhibitors (acalabrutinib and ibrutinib) decreased the duration of mechanical ventilation and mortality rate for hospitalized patients with severe COVID-19. Herein, we review the current information regarding the role of BTK in severe acute respiratory syndrome coronavirus 2 infections and the suitability of its inhibitors as drugs to treat COVID-19. The use of BTK inhibitors in the management of COVID-19 shows promise in reducing the severity of the immune response to the infection and thus mortality. However, BTK inhibition may be contributing in other ways to inhibit the effects of the virus and this will need to be carefully studied.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Adenina/análogos & derivados , Adenina/farmacología , Agammaglobulinemia Tirosina Quinasa/metabolismo , Antivirales/efectos adversos , Benzamidas/farmacología , COVID-19/complicaciones , COVID-19/enzimología , Humanos , Pulmón/efectos de los fármacos , Pulmón/virología , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/virología , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , Trombosis/tratamiento farmacológico , Trombosis/virología
15.
ChemMedChem ; 16(2): 340-354, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: covidwho-1044678

RESUMEN

Inhibition of coronavirus (CoV)-encoded papain-like cysteine proteases (PLpro ) represents an attractive strategy to treat infections by these important human pathogens. Herein we report on structure-activity relationships (SAR) of the noncovalent active-site directed inhibitor (R)-5-amino-2-methyl-N-(1-(naphthalen-1-yl)ethyl) benzamide (2 b), which is known to bind into the S3 and S4 pockets of the SARS-CoV PLpro . Moreover, we report the discovery of isoindolines as a new class of potent PLpro inhibitors. The studies also provide a deeper understanding of the binding modes of this inhibitor class. Importantly, the inhibitors were also confirmed to inhibit SARS-CoV-2 replication in cell culture suggesting that, due to the high structural similarities of the target proteases, inhibitors identified against SARS-CoV PLpro are valuable starting points for the development of new pan-coronaviral inhibitors.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , Proteasas 3C de Coronavirus/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Isoindoles/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/síntesis química , Antivirales/metabolismo , Benzamidas/síntesis química , Benzamidas/metabolismo , Dominio Catalítico , Chlorocebus aethiops , Proteasas 3C de Coronavirus/química , Cristalografía por Rayos X , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/metabolismo , Isoindoles/síntesis química , Isoindoles/metabolismo , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Relación Estructura-Actividad , Células Vero , Replicación Viral/efectos de los fármacos
16.
Eur J Pharmacol ; 892: 173779, 2021 Feb 05.
Artículo en Inglés | MEDLINE | ID: covidwho-959761

RESUMEN

The rapid outbreak of the COVID-19 also known as SARS-CoV2 has been declared pandemic with serious global concern. As there is no effective therapeutic against COVID-19, there is an urgent need for explicit treatment against it. The focused objective of the current study is to propose promising drug candidates against the newly identified potential therapeutic target (endonuclease, NSP15) of SARS-CoV2. NSP15 is an attractive druggable target due to its critical role in SARS-CoV2 replication and virulence in addition to interference with the host immune system. Here in the present study, we integrated the high throughput computational screening and dynamic simulation approach to identify the most promising candidate lead compound against NSP15.5-fluoro-2-oxo-1H-pyrazine-3-carboxamide (favipiravir), (3R,4R, 5R)-3,4-Bis(benzyloxy)-5-((benzyloxy) methyl) dihydrofuran-2(3H)-one) remedesivir, 1,3-thiazol-5-ylmethyl N-[(2S,3S, 5S)-3-hydroxy-5-[[(2 S)-3-methyl-2-[[methyl-[(2-propan-2-yl-1,3-thiazol-4-yl)methyl]carbamoyl]amino]butanoyl]amino]-1,6-diphenylhexan-2-yl]carbamate (ritonavir), ethyl (3R,4R, 5S)-4-acetamido-5-amino-3-pentan-3-yloxycyclohexene-1-carboxylate (oseltamivir), and (2 S)-N-[(2S,4S, 5S)-5-[[2-(2,6-dimethylphenoxy)acetyl]amino]-4-hydroxy-1,6-diphenylhexan-2-yl]-3-methyl-2-(2-oxo-1,3-diazinan-1-yl)butanamide (lopinavir) were chosen as a training set to generate the pharmacophore model. A dataset of ~140,000 compounds library was screened against the designed pharmacophore model and 10 unique compounds were selected that passed successfully through geometry constraints, Lipinski Rule of 5, and ADME/Tox filters along with a strong binding affinity for NSP15 binding cavity. The best fit compound was selected for dynamic simulation to have detailed structural features critical for binding with the NSP15 protein. Given our detailed integrative computational analysis, a Small molecule (3,3-Dimethyl-N-[4-(1-piperidinylcarbonyl) phenyl] butanamide) with drug-like properties and high binding affinity with the NSP15 is proposed as a most promising potential drug against COVID-19. The current computational integrative approach may complement high-throughput screening and the shortlisted small molecule may contribute to selective targeting of NSP15 to stop the replication of SARS-CoV2.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , COVID-19/metabolismo , Endorribonucleasas/metabolismo , Piperidinas/farmacología , SARS-CoV-2 , Proteínas no Estructurales Virales/metabolismo , Antivirales/farmacocinética , Antivirales/toxicidad , Benzamidas/farmacocinética , Diseño de Fármacos , Endorribonucleasas/química , Ensayos Analíticos de Alto Rendimiento , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Piperidinas/farmacocinética , Proteínas no Estructurales Virales/química
17.
Pulm Pharmacol Ther ; 66: 101978, 2021 02.
Artículo en Inglés | MEDLINE | ID: covidwho-947382

RESUMEN

The recent pandemic of COVID-19 caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presents an extraordinary challenge to identify effective drugs for prevention and treatment. The pathogenesis implicate acute respiratory disorder (ARD) which is attributed to significantly triggered "cytokine storm" and compromised immune system. This article summarizes the likely benefits of roflumilast, a Phosphodiesterase-4 (PDE-4) inhibitor as a comprehensive support COVID-19 pathogenesis. Roflumilast, a well-known anti-inflammatory and immunomodulatory drug, is protective against respiratory models of chemical and smoke induced lung damage. There is significant data which demonstrate the protective effect of PDE-4 inhibitor in respiratory viral models and is likely to be beneficial in combating COVID-19 pathogenesis. Roflumilast is effective in patients with severe COPD by reducing the rate of exacerbations with the improvement of the lung function, which might further be beneficial for better clinical outcomes in COVID-19 patients. However, further clinical trials are warranted to examine this conjecture.


Asunto(s)
Aminopiridinas/uso terapéutico , Antiinflamatorios/uso terapéutico , Benzamidas/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Aminopiridinas/efectos adversos , Aminopiridinas/farmacología , Antiinflamatorios/efectos adversos , Antiinflamatorios/farmacología , Benzamidas/efectos adversos , Benzamidas/farmacología , COVID-19/inmunología , Ciclopropanos/efectos adversos , Ciclopropanos/farmacología , Ciclopropanos/uso terapéutico , Citocinas/biosíntesis , Mediadores de Inflamación/metabolismo , Pandemias , Inhibidores de Fosfodiesterasa 4/efectos adversos , Inhibidores de Fosfodiesterasa 4/farmacología
18.
Eur J Pharmacol ; 889: 173615, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: covidwho-808499

RESUMEN

Nowadays, coronavirus disease 2019 (COVID-19) represents the most serious inflammatory respiratory disease worldwide. Despite many proposed therapies, no effective medication has yet been approved. Neutrophils appear to be the key mediator for COVID-19-associated inflammatory immunopathologic, thromboembolic and fibrotic complications. Thus, for any therapeutic agent to be effective, it should greatly block the neutrophilic component of COVID-19. One of the effective therapeutic approaches investigated to reduce neutrophil-associated inflammatory lung diseases with few adverse effects was roflumilast. Being a highly selective phosphodiesterase-4 inhibitors (PDE4i), roflumilast acts by enhancing the level of cyclic adenosine monophosphate (cAMP), that probably potentiates its anti-inflammatory action via increasing neprilysin (NEP) activity. Because activating NEP was previously reported to mitigate several airway inflammatory ailments; this review thoroughly discusses the proposed NEP-based therapeutic properties of roflumilast, which may be of great importance in curing COVID-19. However, further clinical studies are required to confirm this strategy and to evaluate its in vivo preventive and therapeutic efficacy against COVID-19.


Asunto(s)
Aminopiridinas/farmacología , Aminopiridinas/uso terapéutico , Benzamidas/farmacología , Benzamidas/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Neprilisina/efectos de los fármacos , Ciclopropanos/farmacología , Ciclopropanos/uso terapéutico , Humanos , Pandemias , SARS-CoV-2
19.
Microbiol Immunol ; 64(9): 635-639, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: covidwho-613452

RESUMEN

In this study, the anti-severe acute respiratory syndrome coronavirus-2 (anti-SARS-CoV-2) activity of mycophenolic acid (MPA) and IMD-0354 was analyzed. These compounds were chosen based on their antiviral activities against other coronaviruses. Because they also inhibit dengue virus (DENV) infection, other anti-DENV compounds/drugs were also assessed. On SARS-CoV-2-infected VeroE6/TMPRSS2 monolayers, both MPA and IMD-0354, but not other anti-DENV compounds/drugs, showed significant anti-SARS-CoV-2 activity. Although MPA reduced the viral RNA level by only approximately 100-fold, its half maximal effective concentration was as low as 0.87 µ m, which is easily achievable at therapeutic doses of mycophenolate mofetil. MPA targets the coronaviral papain-like protease and an in-depth study on its mechanism of action would be useful in the development of novel anti-SARS-CoV-2 drugs.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Ácido Micofenólico/farmacología , Neumonía Viral/tratamiento farmacológico , Animales , COVID-19 , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Virus del Dengue/efectos de los fármacos , Humanos , Pandemias , Neumonía Viral/virología , SARS-CoV-2 , Células Vero , Replicación Viral/efectos de los fármacos
20.
Sci Immunol ; 5(48)2020 06 05.
Artículo en Inglés | MEDLINE | ID: covidwho-545978

RESUMEN

Patients with severe COVID-19 have a hyperinflammatory immune response suggestive of macrophage activation. Bruton tyrosine kinase (BTK) regulates macrophage signaling and activation. Acalabrutinib, a selective BTK inhibitor, was administered off-label to 19 patients hospitalized with severe COVID-19 (11 on supplemental oxygen; 8 on mechanical ventilation), 18 of whom had increasing oxygen requirements at baseline. Over a 10-14 day treatment course, acalabrutinib improved oxygenation in a majority of patients, often within 1-3 days, and had no discernable toxicity. Measures of inflammation - C-reactive protein and IL-6 - normalized quickly in most patients, as did lymphopenia, in correlation with improved oxygenation. At the end of acalabrutinib treatment, 8/11 (72.7%) patients in the supplemental oxygen cohort had been discharged on room air, and 4/8 (50%) patients in the mechanical ventilation cohort had been successfully extubated, with 2/8 (25%) discharged on room air. Ex vivo analysis revealed significantly elevated BTK activity, as evidenced by autophosphorylation, and increased IL-6 production in blood monocytes from patients with severe COVID-19 compared with blood monocytes from healthy volunteers. These results suggest that targeting excessive host inflammation with a BTK inhibitor is a therapeutic strategy in severe COVID-19 and has led to a confirmatory international prospective randomized controlled clinical trial.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Benzamidas/farmacología , Benzamidas/uso terapéutico , Betacoronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Pirazinas/farmacología , Pirazinas/uso terapéutico , Agammaglobulinemia Tirosina Quinasa/metabolismo , Anciano , Anciano de 80 o más Años , COVID-19 , Infecciones por Coronavirus/virología , Enfermedad Crítica , Femenino , Estudios de Seguimiento , Humanos , Inflamación/tratamiento farmacológico , Inflamación/virología , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Pandemias , Neumonía Viral/virología , Estudios Prospectivos , Respiración Artificial , SARS-CoV-2 , Resultado del Tratamiento , Tratamiento Farmacológico de COVID-19
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA