Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Virol ; 96(22): e0099722, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: covidwho-2097918

RESUMEN

Modification of the hepatitis C virus (HCV) positive-strand RNA genome by N6-methyladenosine (m6A) regulates the viral life cycle. This life cycle takes place solely in the cytoplasm, while m6A addition on cellular mRNA takes place in the nucleus. Thus, the mechanisms by which m6A is deposited on the viral RNA have been unclear. In this work, we find that m6A modification of HCV RNA by the m6A-methyltransferase proteins methyltransferase-like 3 and 14 (METTL3 and METTL14) is regulated by Wilms' tumor 1-associating protein (WTAP). WTAP, a predominantly nuclear protein, is an essential member of the cellular mRNA m6A-methyltransferase complex and known to target METTL3 to mRNA. We found that HCV infection induces localization of WTAP to the cytoplasm. Importantly, we found that WTAP is required for both METTL3 interaction with HCV RNA and m6A modification across the viral RNA genome. Further, we found that WTAP, like METTL3 and METTL14, negatively regulates the production of infectious HCV virions, a process that we have previously shown is regulated by m6A. Excitingly, WTAP regulation of both HCV RNA m6A modification and virion production was independent of its ability to localize to the nucleus. Together, these results reveal that WTAP is critical for HCV RNA m6A modification by METTL3 and METTL14 in the cytoplasm. IMPORTANCE Positive-strand RNA viruses such as HCV represent a significant global health burden. Previous work has described that HCV RNA contains the RNA modification m6A and how this modification regulates viral infection. Yet, how this modification is targeted to HCV RNA has remained unclear due to the incompatibility of the nuclear cellular processes that drive m6A modification with the cytoplasmic HCV life cycle. In this study, we present evidence for how m6A modification is targeted to HCV RNA in the cytoplasm by a mechanism in which WTAP recruits the m6A-methyltransferase METTL3 to HCV RNA. This targeting strategy for m6A modification of cytoplasmic RNA viruses is likely relevant for other m6A-modified positive-strand RNA viruses with cytoplasmic life cycles such as enterovirus 71 and SARS-CoV-2 and provides an exciting new target for potential antiviral therapies.


Asunto(s)
Proteínas de Ciclo Celular , Hepatitis C , Metiltransferasas , Factores de Empalme de ARN , Humanos , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatitis C/genética , Hepatitis C/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Factores de Empalme de ARN/metabolismo , ARN Mensajero/genética , ARN Viral/genética , ARN Viral/metabolismo
2.
Nucleic Acids Res ; 50(15): 8700-8718, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: covidwho-1973223

RESUMEN

FACT (FAcilitates Chromatin Transcription) is a heterodimeric protein complex composed of SUPT16H and SSRP1, and a histone chaperone participating in chromatin remodeling during gene transcription. FACT complex is profoundly regulated, and contributes to both gene activation and suppression. Here we reported that SUPT16H, a subunit of FACT, is acetylated in both epithelial and natural killer (NK) cells. The histone acetyltransferase TIP60 contributes to the acetylation of SUPT16H middle domain (MD) at lysine 674 (K674). Such acetylation of SUPT16H is recognized by bromodomain protein BRD4, which promotes protein stability of SUPT16H in both epithelial and NK cells. We further demonstrated that SUPT16H-BRD4 associates with histone modification enzymes (HDAC1, EZH2), and further regulates their activation status and/or promoter association as well as affects the relevant histone marks (H3ac, H3K9me3 and H3K27me3). BRD4 is known to profoundly regulate interferon (IFN) signaling, while such function of SUPT16H has never been explored. Surprisingly, our results revealed that SUPT16H genetic knockdown via RNAi or pharmacological inhibition by using its inhibitor, curaxin 137 (CBL0137), results in the induction of IFNs and interferon-stimulated genes (ISGs). Through this mechanism, depletion or inhibition of SUPT16H is shown to efficiently inhibit infection of multiple viruses, including Zika, influenza, and SARS-CoV-2. Furthermore, we demonstrated that depletion or inhibition of SUPT16H also causes the remarkable activation of IFN signaling in NK cells, which promotes the NK-mediated killing of virus-infected cells in a co-culture system using human primary NK cells. Overall, our studies unraveled the previously un-appreciated role of FACT complex in coordinating with BRD4 and regulating IFN signaling in both epithelial and NK cells, and also proposed the novel application of the FACT inhibitor CBL0137 to treat viral infections.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Células Epiteliales/metabolismo , Interferones/metabolismo , Células Asesinas Naturales/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , COVID-19 , Proteínas de Unión al ADN/genética , Células Epiteliales/inmunología , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Células Asesinas Naturales/inmunología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , SARS-CoV-2 , Factores de Elongación Transcripcional/genética , Virus Zika/metabolismo , Infección por el Virus Zika
3.
Structure ; 30(9): 1224-1232.e5, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1895449

RESUMEN

Emerging new variants of SARS-CoV-2 and inevitable acquired drug resistance call for the continued search of new pharmacological targets to fight the potentially fatal infection. Here, we describe the mechanisms by which the E protein of SARS-CoV-2 hijacks the human transcriptional regulator BRD4. We found that SARS-CoV-2 E is acetylated in vivo and co-immunoprecipitates with BRD4 in human cells. Bromodomains (BDs) of BRD4 bind to the C-terminus of the E protein, acetylated by human acetyltransferase p300, whereas the ET domain of BRD4 recognizes the unmodified motif of the E protein. Inhibitors of BRD4 BDs, JQ1 or OTX015, decrease SARS-CoV-2 infectivity in lung bronchial epithelial cells, indicating that the acetyllysine binding function of BDs is necessary for the virus fitness and that BRD4 represents a potential anti-COVID-19 target. Our findings provide insight into molecular mechanisms that contribute to SARS-CoV-2 pathogenesis and shed light on a new strategy to block SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Proteínas de Ciclo Celular/metabolismo , Proteínas de la Envoltura de Coronavirus/metabolismo , SARS-CoV-2/fisiología , Factores de Transcripción/metabolismo , COVID-19/virología , Humanos , Proteínas Nucleares/metabolismo , Unión Proteica , Dominios Proteicos
4.
Biomed Pharmacother ; 152: 113230, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: covidwho-1881709

RESUMEN

BACKGROUND: Bromodomain and extraterminal proteins (BETs) are more than just epigenetic regulators of transcription. Here we highlight a new role for the BET protein BRD4 in the maintenance of higher order chromatin structure at Topologically Associating Domain Boundaries (TADBs). BD2-selective and pan (non-selective) BET inhibitors (BETi) differentially support chromatin structure, selectively affecting transcription and cell viability. METHODS: Using RNA-seq and BRD4 ChIP-seq, the differential effect of BETi treatment on the transcriptome and BRD4 chromatin occupancy of human aortic endothelial cells from diabetic patients (dHAECs) stimulated with TNFα was evaluated. Chromatin decondensation and DNA fragmentation was assessed by immunofluorescence imaging and quantification. Key dHAEC findings were verified in proliferating monocyte-like THP-1 cells using real time-PCR, BRD4 co-immunoprecipitation studies, western blots, proliferation and apoptosis assays. FINDINGS: We discovered that 1) BRD4 co-localizes with Ying-Yang 1 (YY1) at TADBs, critical chromatin structure complexes proximal to many DNA repair genes. 2) BD2-selective BETi enrich BRD4/YY1 associations, while pan-BETi do not. 3) Failure to support chromatin structures through BRD4/YY1 enrichment inhibits DNA repair gene transcription, which induces DNA damage responses, and causes widespread chromatin decondensation, DNA fragmentation, and apoptosis. 4) BD2-selective BETi maintain high order chromatin structure and cell viability, while reducing deleterious pro-inflammatory transcription. INTERPRETATION: BRD4 plays a previously unrecognized role at TADBs. BETi differentially impact TADB stability. Our results provide translational insight for the development of BETi as therapeutics for a range of diseases including CVD, chronic kidney disease, cancer, and COVID-19.


Asunto(s)
COVID-19 , Factores de Transcripción , Proteínas de Ciclo Celular/metabolismo , Cromatina , Células Endoteliales/metabolismo , Epigénesis Genética , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo
5.
Biochem Biophys Res Commun ; 616: 14-18, 2022 08 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1850694

RESUMEN

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a major threat to human health. As a unique putative protein of SARS-CoV-2, the N-terminus of ORF10 can be recognized by ZYG11B, a substrate receptor of the Cullin 2-RING E3 ubiquitin ligase (CRL2). Here we elucidated recognition mechanism of ORF10 N-terminus by ZYG11B through presenting the crystal structure of ZYG11B bound to ORF10 N-terminal peptide. Our work expands the current understanding of ORF10 interaction with ZYG11B, and may also inspire the development of novel therapies for COVID-19.


Asunto(s)
COVID-19 , Proteínas de Ciclo Celular , Sistemas de Lectura Abierta , Ubiquitina-Proteína Ligasas , COVID-19/metabolismo , COVID-19/virología , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas Cullin , Humanos , SARS-CoV-2/química , SARS-CoV-2/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo
6.
Int J Biol Sci ; 17(12): 3224-3238, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1524470

RESUMEN

Mechanisms of breast cancer progression and invasion, often involve alteration of hormonal signaling, and upregulation and/or activation of signal transduction pathways that input to cell cycle regulation. Herein, we describe a rationally designed first-in-class novel small molecule inhibitor for targeting oncogenic and hormonal signaling in ER-positive breast cancer. BC-N102 treatment exhibits dose-dependent cytotoxic effects against ER+ breast cancer cell lines. BC-N102 exhibited time course- and dose-dependent cell cycle arrest via downregulation of the estrogen receptor (ER), progesterone receptor (PR), androgen receptor (AR), phosphatidylinositol 3-kinase (PI3K), phosphorylated (p)-extracellular signal-regulated kinase (ERK), p-Akt, CDK2, and CDK4 while increasing p38 mitogen-activated protein kinase (MAPK), and mineralocorticoid receptor (MR) signaling in breast cancer cell line. In addition, we found that BC-N102 suppressed breast cancer tumorigenesis in vivo and prolonged the survival of animals. Our results suggest that the proper application of BC-N102 may be a beneficial chemotherapeutic strategy for ER+ breast cancer patients.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Fase G1/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Animales , Biomarcadores de Tumor/genética , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , División Celular , Línea Celular Tumoral , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/genética , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Dosis Máxima Tolerada , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Virol ; 95(17): e0079421, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1350003

RESUMEN

Increased mortality in COVID-19 cases is often associated with microvascular complications. We have recently shown that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein promotes an inflammatory cytokine interleukin 6 (IL-6)/IL-6R-induced trans signaling response and alarmin secretion. Virus-infected or spike-transfected human epithelial cells exhibited an increase in senescence, with a release of senescence-associated secretory phenotype (SASP)-related inflammatory molecules. Introduction of the bromodomain-containing protein 4 (BRD4) inhibitor AZD5153 to senescent epithelial cells reversed this effect and reduced SASP-related inflammatory molecule release in TMNK-1 or EAhy926 (representative human endothelial cell lines), when cells were exposed to cell culture medium (CM) derived from A549 cells expressing SARS-CoV-2 spike protein. Cells also exhibited a senescence phenotype with enhanced p16, p21, and senescence-associated ß-galactosidase (SA-ß-Gal) expression and triggered SASP pathways. Inhibition of IL-6 trans signaling by tocilizumab and inhibition of inflammatory receptor signaling by the Bruton's tyrosine kinase (BTK) inhibitor zanubrutinib, prior to exposure of CM to endothelial cells, inhibited p21 and p16 induction. We also observed an increase in reactive oxygen species (ROS) in A549 spike-transfected and endothelial cells exposed to spike-transfected CM. ROS generation in endothelial cell lines was reduced after treatment with tocilizumab and zanubrutinib. Cellular senescence was associated with an increased level of the endothelial adhesion molecules vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1), which have in vitro leukocyte attachment potential. Inhibition of senescence or SASP function prevented VCAM-1/ICAM-1 expression and leukocyte attachment. Taken together, we identified that human endothelial cells exposed to cell culture supernatant derived from SARS-CoV-2 spike protein expression displayed cellular senescence markers, leading to enhanced leukocyte adhesion. IMPORTANCE The present study was aimed at examining the underlying mechanism of extrapulmonary manifestations of SARS-CoV-2 spike protein-associated pathogenesis, with the notion that infection of the pulmonary epithelium can lead to mediators that drive endothelial dysfunction. We utilized SARS-CoV-2 spike protein expression in cultured human hepatocytes (Huh7.5) and pneumocytes (A549) to generate conditioned culture medium (CM). Endothelial cell lines (TMNK-1 or EAhy926) treated with CM exhibited an increase in cellular senescence markers by a paracrine mode and led to leukocyte adhesion. Overall, the link between these responses in endothelial cell senescence and a potential contribution to microvascular complication in productively SARS-CoV-2-infected humans is implicated. Furthermore, the use of inhibitors (BTK, IL-6, and BRD4) showed a reverse effect in the senescent cells. These results may support the selection of potential adjunct therapeutic modalities to impede SARS-CoV-2-associated pathogenesis.


Asunto(s)
Senescencia Celular , Células Endoteliales/metabolismo , Leucocitos/metabolismo , Comunicación Paracrina , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células A549 , Adhesión Celular , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Células Endoteliales/patología , Células Endoteliales/virología , Compuestos Heterocíclicos con 2 Anillos/farmacología , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/metabolismo , Leucocitos/patología , Leucocitos/virología , Piperazinas/farmacología , Pirazoles , Piridazinas , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-6/metabolismo , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
8.
Biomolecules ; 11(8)2021 07 30.
Artículo en Inglés | MEDLINE | ID: covidwho-1334993

RESUMEN

The recent pandemic we are experiencing caused by the coronavirus disease 2019 (COVID-19) has put the world's population on the rack, with more than 191 million cases and more than 4.1 million deaths confirmed to date. This disease is caused by a new type of coronavirus, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A massive proteomic analysis has revealed that one of the structural proteins of the virus, the E protein, interacts with BRD2 and BRD4 proteins of the Bromodomain and Extra Terminal domain (BET) family of proteins. BETs are essential to cell cycle progression, inflammation and immune response and have also been strongly associated with infection by different types of viruses. The fundamental role BET proteins play in transcription makes them appropriate targets for the propagation strategies of some viruses. Recognition of histone acetylation by BET bromodomains is essential for transcription control. The development of drugs mimicking acetyl groups, and thereby able to displace BET proteins from chromatin, has boosted interest on BETs as attractive targets for therapeutic intervention. The success of these drugs against a variety of diseases in cellular and animal models has been recently enlarged with promising results from SARS-CoV-2 infection studies.


Asunto(s)
COVID-19/metabolismo , Proteínas de Ciclo Celular/metabolismo , Factores de Transcripción/metabolismo , COVID-19/inmunología , COVID-19/virología , Proteínas de la Envoltura de Coronavirus/metabolismo , Humanos , Inmunidad Innata , Unión Proteica
9.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artículo en Inglés | MEDLINE | ID: covidwho-1172591

RESUMEN

In order to understand the transmission and virulence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), it is necessary to understand the functions of each of the gene products encoded in the viral genome. One feature of the SARS-CoV-2 genome that is not present in related, common coronaviruses is ORF10, a putative 38-amino acid protein-coding gene. Proteomic studies found that ORF10 binds to an E3 ubiquitin ligase containing Cullin-2, Rbx1, Elongin B, Elongin C, and ZYG11B (CRL2ZYG11B). Since CRL2ZYG11B mediates protein degradation, one possible role for ORF10 is to "hijack" CRL2ZYG11B in order to target cellular, antiviral proteins for ubiquitylation and subsequent proteasomal degradation. Here, we investigated whether ORF10 hijacks CRL2ZYG11B or functions in other ways, for example, as an inhibitor or substrate of CRL2ZYG11B While we confirm the ORF10-ZYG11B interaction and show that the N terminus of ORF10 is critical for it, we find no evidence that ORF10 is functioning to inhibit or hijack CRL2ZYG11B Furthermore, ZYG11B and its paralog ZER1 are dispensable for SARS-CoV-2 infection in cultured cells. We conclude that the interaction between ORF10 and CRL2ZYG11B is not relevant for SARS-CoV-2 infection in vitro.


Asunto(s)
COVID-19/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cullin/metabolismo , Complejos Multiproteicos/metabolismo , Sistemas de Lectura Abierta , SARS-CoV-2/metabolismo , Proteínas Virales/metabolismo , COVID-19/genética , Proteínas de Ciclo Celular/genética , Proteínas Cullin/genética , Células HEK293 , Humanos , Complejos Multiproteicos/genética , SARS-CoV-2/genética , Proteínas Virales/genética
10.
Cell ; 184(8): 2167-2182.e22, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: covidwho-1135274

RESUMEN

Cardiac injury and dysfunction occur in COVID-19 patients and increase the risk of mortality. Causes are ill defined but could be through direct cardiac infection and/or inflammation-induced dysfunction. To identify mechanisms and cardio-protective drugs, we use a state-of-the-art pipeline combining human cardiac organoids with phosphoproteomics and single nuclei RNA sequencing. We identify an inflammatory "cytokine-storm", a cocktail of interferon gamma, interleukin 1ß, and poly(I:C), induced diastolic dysfunction. Bromodomain-containing protein 4 is activated along with a viral response that is consistent in both human cardiac organoids (hCOs) and hearts of SARS-CoV-2-infected K18-hACE2 mice. Bromodomain and extraterminal family inhibitors (BETi) recover dysfunction in hCOs and completely prevent cardiac dysfunction and death in a mouse cytokine-storm model. Additionally, BETi decreases transcription of genes in the viral response, decreases ACE2 expression, and reduces SARS-CoV-2 infection of cardiomyocytes. Together, BETi, including the Food and Drug Administration (FDA) breakthrough designated drug, apabetalone, are promising candidates to prevent COVID-19 mediated cardiac damage.


Asunto(s)
COVID-19/complicaciones , Cardiotónicos/uso terapéutico , Proteínas de Ciclo Celular/antagonistas & inhibidores , Cardiopatías/tratamiento farmacológico , Quinazolinonas/uso terapéutico , Factores de Transcripción/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Citocinas/metabolismo , Femenino , Cardiopatías/etiología , Células Madre Embrionarias Humanas , Humanos , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/metabolismo , Tratamiento Farmacológico de COVID-19
11.
Drug Discov Today ; 26(5): 1302-1310, 2021 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1077854

RESUMEN

The synthetic antibacterial drug clofoctol (CFT) has long been used to treat respiratory tract infections in Europe. In recent years, the drug was found to target two biologically important proteins, the Cdc7/Dbf4 protein kinase complex and the mRNA-binding protein cold shock domain containing E1 (CSDE1), also known as upstream-of-N-Ras protein (UNR). These interactions are at the origin of the antitumor activity of CFT, recently evidenced in prostate cancer and neuroglioma. Drug-protein binding models provide a structural basis to guide the design of more potent anticancer compounds. A renewed interest in CFT can be anticipated for the treatment of cancers, and possibly Coronavirus 2019 (COVID-19).


Asunto(s)
Antibacterianos/uso terapéutico , Antineoplásicos/uso terapéutico , Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos , Neoplasias/tratamiento farmacológico , Animales , Antibacterianos/efectos adversos , Antineoplásicos/efectos adversos , Antivirales/efectos adversos , COVID-19/metabolismo , COVID-19/virología , Proteínas de Ciclo Celular/metabolismo , Clorobencenos , Cresoles/efectos adversos , Cresoles/uso terapéutico , Proteínas de Unión al ADN/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al ARN/metabolismo
12.
Virologie (Montrouge) ; 24(6): 381-418, 2020 12 01.
Artículo en Francés | MEDLINE | ID: covidwho-1030336

RESUMEN

The innate immune response is nonspecific and constitutes the first line of defense against infections by pathogens, mainly by enabling their elimination by phagocytosis or apoptosis. In immune cells, this response is characterized, amongst others, by the synthesis of restriction factors, a class of proteins whose role is to inhibit viral replication. Among them, the proteins of the APOBEC3 (Apolipoprotein B mRNA-editing Enzyme Catalytic polypeptide-like 3 or A3) family are major antiviral factors that target a wide range of viruses. One of their targets is the Human Immunodeficiency Virus Type 1 (HIV-1): the deaminase activity of some A3 proteins converts a fraction of cytidines of the viral genome into uridines, impairing its expression. Nevertheless, HIV-1 counteracts A3 proteins thanks to its Vif protein, which inhibits them by hijacking several cellular mechanisms. Besides, APOBEC3 proteins help maintaining the genome integrity by inhibiting retroelements but they also contribute to carcinogenesis, as it is the case for A3A and A3B, two major factors in this process. The large range of A3 activities, combined with recent studies showing their implication in the regulation of emerging viruses (Zika, SARS-CoV-2), allow A3 and their viral partners to be considered as therapeutic areas.


Asunto(s)
Desaminasas APOBEC/fisiología , COVID-19/inmunología , Inmunidad Innata , Adulto , Secuencias de Aminoácidos , Animales , Proteínas de Ciclo Celular/metabolismo , Citidina Desaminasa/fisiología , Reparación del ADN , ADN Viral/metabolismo , Desaminación , Humanos , Mamíferos/metabolismo , MicroARNs/genética , Modelos Moleculares , Terapia Molecular Dirigida , Mutagénesis , Neoplasias/enzimología , Neoplasias/etiología , Neoplasias/genética , Pronóstico , Conformación Proteica , Edición de ARN , Relación Estructura-Actividad , Transcripción Genética , Proteínas Virales/metabolismo , Virosis/tratamiento farmacológico , Virosis/enzimología , Virosis/inmunología , Replicación Viral
13.
SLAS Discov ; 26(3): 336-344, 2021 03.
Artículo en Inglés | MEDLINE | ID: covidwho-934236

RESUMEN

The reuse of preexisting small molecules for a novel emerging disease threat is a rapid measure to discover unknown applications for previously validated therapies. A pertinent and recent example where such a strategy could be employed is in the fight against coronavirus disease 2019 (COVID-19). Therapies designed or discovered to target viral proteins also have off-target effects on the host proteome when employed in a complex physiological environment. This study aims to assess these host cell targets for a panel of FDA-approved antiviral compounds including remdesivir, using the cellular thermal shift assay (CETSA) coupled with mass spectrometry (CETSA MS) in noninfected cells. CETSA MS is a powerful method to delineate direct and indirect interactions between small molecules and protein targets in intact cells. Biologically active compounds can induce changes in thermal stability, in their primary binding partners, and in proteins that in turn interact with the direct targets. Such engagement of host targets by antiviral drugs may contribute to the clinical effect against the virus but can also constitute a liability. We present here a comparative study of CETSA molecular target engagement fingerprints of antiviral drugs to better understand the link between off-targets and efficacy.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Antivirales/farmacología , Proteínas de Ciclo Celular/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Adenosina/análogos & derivados , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Reposicionamiento de Medicamentos , Furanos/farmacología , Células Hep G2 , Humanos , Espectrometría de Masas , Proteómica/métodos , Pirroles/farmacología , Triazinas/farmacología , Estados Unidos , United States Food and Drug Administration , Tratamiento Farmacológico de COVID-19
14.
Proc Natl Acad Sci U S A ; 117(29): 17195-17203, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: covidwho-624792

RESUMEN

The vast majority of intracellular protein targets are refractory toward small-molecule therapeutic engagement, and additional therapeutic modalities are needed to overcome this deficiency. Here, the identification and characterization of a natural product, WDB002, reveals a therapeutic modality that dramatically expands the currently accepted limits of druggability. WDB002, in complex with the FK506-binding protein (FKBP12), potently and selectively binds the human centrosomal protein 250 (CEP250), resulting in disruption of CEP250 function in cells. The recognition mode is unprecedented in that the targeted domain of CEP250 is a coiled coil and is topologically featureless, embodying both a structural motif and surface topology previously considered on the extreme limits of "undruggability" for an intracellular target. Structural studies reveal extensive protein-WDB002 and protein-protein contacts, with the latter being distinct from those seen in FKBP12 ternary complexes formed by FK506 and rapamycin. Outward-facing structural changes in a bound small molecule can thus reprogram FKBP12 to engage diverse, otherwise "undruggable" targets. The flat-targeting modality demonstrated here has the potential to expand the druggable target range of small-molecule therapeutics. As CEP250 was recently found to be an interaction partner with the Nsp13 protein of the SARS-CoV-2 virus that causes COVID-19 disease, it is possible that WDB002 or an analog may exert useful antiviral activity through its ability to form high-affinity ternary complexes containing CEP250 and FKBP12.


Asunto(s)
Actinobacteria/genética , Antivirales/farmacología , Genoma Bacteriano , Macrólidos/farmacología , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína 1A de Unión a Tacrolimus/química , Proteína 1A de Unión a Tacrolimus/metabolismo , Actinobacteria/metabolismo , Secuencia de Aminoácidos , Antivirales/química , Antivirales/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Calcineurina/genética , Calcineurina/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Evolución Molecular , Células HEK293 , Humanos , Macrólidos/química , Macrólidos/metabolismo , Modelos Moleculares , Conformación Proteica , Homología de Secuencia , Sirolimus/química , Sirolimus/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
15.
Mol Cell Endocrinol ; 515: 110917, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: covidwho-661768

RESUMEN

Obesity patients are more susceptible to develop COVID-19 severe outcome due to the role of angiotensin-converting enzyme 2 (ACE2) in the viral infection. ACE2 is regulated in the human cells by different genes associated with increased (TLR3, HAT1, HDAC2, KDM5B, SIRT1, RAB1A, FURIN and ADAM10) or decreased (TRIB3) virus replication. RNA-seq data revealed 14857 genes expressed in human subcutaneous adipocytes, including genes mentioned above. Irisin treatment increased by 3-fold the levels of TRIB3 transcript and decreased the levels of other genes. The decrease in FURIN and ADAM10 expression enriched diverse biological processes, including extracellular structure organization. Our results, in human subcutaneous adipocytes cell culture, indicate a positive effect of irisin on the expression of multiple genes related to viral infection by SARS-CoV-2; furthermore, translatable for other tissues and organs targeted by the novel coronavirus and present, thus, promising approaches for the treatment of COVID-19 infection as therapeutic strategy to decrease ACE2 regulatory genes.


Asunto(s)
Adipocitos/efectos de los fármacos , Fibronectinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína ADAM10/genética , Proteína ADAM10/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Enzima Convertidora de Angiotensina 2 , Betacoronavirus/genética , Betacoronavirus/metabolismo , COVID-19 , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Infecciones por Coronavirus/virología , Fibronectinas/genética , Fibronectinas/metabolismo , Furina/genética , Furina/metabolismo , Ontología de Genes , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Anotación de Secuencia Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Obesidad/virología , Pandemias , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/virología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , SARS-CoV-2 , Transducción de Señal , Sirtuina 1/genética , Sirtuina 1/metabolismo , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Proteínas de Unión al GTP rab1/genética , Proteínas de Unión al GTP rab1/metabolismo
16.
J Mol Cell Biol ; 12(12): 946-957, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: covidwho-637628

RESUMEN

For patients with COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the damages to multiple organs have been clinically observed. Since most of current investigations for virus-host interaction are based on cell level, there is an urgent demand to probe tissue-specific features associated with SARS-CoV-2 infection. Based on collected proteomic datasets from human lung, colon, kidney, liver, and heart, we constructed a virus-receptor network, a virus-interaction network, and a virus-perturbation network. In the tissue-specific networks associated with virus-host crosstalk, both common and different key hubs are revealed in diverse tissues. Ubiquitous hubs in multiple tissues such as BRD4 and RIPK1 would be promising drug targets to rescue multi-organ injury and deal with inflammation. Certain tissue-unique hubs such as REEP5 might mediate specific olfactory dysfunction. The present analysis implies that SARS-CoV-2 could affect multi-targets in diverse host tissues, and the treatment of COVID-19 would be a complex task.


Asunto(s)
COVID-19/metabolismo , COVID-19/virología , Interacciones Microbiota-Huesped/fisiología , Proteoma/metabolismo , SARS-CoV-2 , Proteínas de Ciclo Celular/metabolismo , Colon/metabolismo , Colon/virología , Corazón/virología , Humanos , Riñón/metabolismo , Riñón/virología , Hígado/metabolismo , Hígado/virología , Pulmón/metabolismo , Pulmón/virología , Proteínas de la Membrana/metabolismo , Redes y Vías Metabólicas , Miocardio/metabolismo , Pandemias , Mapas de Interacción de Proteínas , Proteómica , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores Virales/metabolismo , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Distribución Tisular , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA