Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Biosens Bioelectron ; 226: 115104, 2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: covidwho-2307021

RESUMEN

The separation of the superimposed electrochemical signals of intracellular guanine (G) and xanthine (X) is difficult, which is great obstacle to the application of cell electrochemistry. In this paper, independent functional modules, G-functional module (G-FM) and X-functional module (X-FM), were constructed by molecular imprinting technology for sensitive detection of G and X without mutual interference, then integrated in dual-functional module cellular electrochemical sensing platform (DMCEP) as signal sensing units. DMCEP transmitted signals of G and X in cells synchronously to two windows by two signal sensing channels, and achieved the separation of superimposed signals of G and X in cells. DMCEP exhibited satisfactory reproducibility with relative standard deviation (RSD) of 3.10 and 2.22 %, repeatability with RSD of 3.72 and 3.05 % for G and X detection, and detection limit 0.05 µΜ for G and 0.06 µΜ for X. Good linear relationships between cell concentrations and the signals of G and X on DMCEP were shown in range of 0.75-85 × 106 and 3-85 × 106 cells/mL, respectively. The growth of MCF-7 cells was tracked by DMCEP, and showed consistent trend with the cell counting method, while the change of cell viability from lag to logarithmic phase captured by DMCEP was earlier than that of cell counting method. This strategy provided the foundation for the establishment of the cell viability electrochemical detection method, and new insights into the simultaneous recording of other analyses with superimposed peak positions and the simultaneous tracking of multiple biomarkers.


Asunto(s)
Técnicas Biosensibles , Guanina , Humanos , Xantina , Guanina/análisis , Reproducibilidad de los Resultados , Células MCF-7 , Técnicas Electroquímicas , Límite de Detección , Electrodos
2.
Anal Chim Acta ; 1252: 341029, 2023 Apr 29.
Artículo en Inglés | MEDLINE | ID: covidwho-2277233

RESUMEN

N-linked glycosylation (N-glycosylation) is a common protein post-translational modification, occurring on more than half of mammalian proteins; in striking contract with small molecule modifications (such as methylation, phosphorylation) with only single structures, N-glycosylation has multiple dimensional structural features (monosaccharide composition, sequence, linkage, anomer), which generates enormous N-glycan structures; and these structures widely regulate protein structure and functions. For the modification site, N-glycosylation occurs on the Asn residue among the consensus N-X-S/T/C (X≠P) motif; mutation-originated amino acid change may lead to loss of such an original motif and thus loss-of-glycosylation (LoG) or gain of such a new motif and thus gain-of-glycosylation (GoG). Both LoG and GoG generates new structures and functions of glycoproteins, which has been observed in the S protein of SARS-Cov-2 as well as malignant diseases. Here we report our glycoproteome-wide qualitative N-glycoproteomics characterization of GoGs in breast cancer Adriamycin drug resistance (ADR) cells (MCF-7/ADR) and cancer stem cells (MCF-7/ADR CSCs); comprehensive N-glycosite and N-glycan structure information at the intact N-glycopeptide level were reported.


Asunto(s)
Adenocarcinoma , COVID-19 , Animales , Humanos , Glicosilación , Células MCF-7 , Glicopéptidos/química , SARS-CoV-2 , Glicoproteínas/química , Polisacáridos , Doxorrubicina , Células Madre Neoplásicas/metabolismo , Mamíferos/metabolismo
3.
Cells ; 11(10)2022 05 19.
Artículo en Inglés | MEDLINE | ID: covidwho-1903275

RESUMEN

BACKGROUND: Breast cancer (BC) is the leading cause of death worldwide. The severity of BC strictly depends on the molecular subtype. The less aggressive hormone-positive subtype is treated with adjuvant endocrine therapy (AET), which causes both physical and psychological side effects. This condition strongly impacts the adherence and persistence of AET among oncologic patients. Moreover, viral infections also constitute a serious problem for public health. Despite their efficacy, antiviral agents present several therapeutic limits. Accordingly, in the present work, we investigated the antitumor and antiviral activities of Orobanche crenata Forssk. (O. crenata), a parasitic plant, endemic to the Mediterranean basin, traditionally known for its beneficial properties for human health. METHODS: The MTT assay was carried out to evaluate the cytotoxic effect of O. crenata leaf extract (OCLE) on human breast cancer cells (MCF-7 and MDA-MB-231) and the primary HFF-1 cell line. The lactic dehydrogenase (LDH) assay was performed on MCF-7 cells to analyze necrotic cell death. The antioxidant effect of OCLE was evaluated by intracellular determination of the reactive oxygen species and thiol groups, by DPPH and ABTS assays. The antiviral activity of OCLE was determined against Poliovirus 1, Echovirus 9, Human respiratory syncytial virus, Adenovirus type 2 and type 5, Coxsackievirus B1 (CoxB1) and B3 (CoxB3), Herpes simplex type 1 (HSV-1) and type 2 (HSV-2), and ß-Coronavirus by the plaque reduction assay. RESULTS: The extract, after 24 h of incubation, did not affect MDA-MB-231 and HFF-1 cell viability. However, at the same time point, it showed a dose-dependent inhibitory effect on MCF-7 cells, with an increase in LDH release. OCLE exhibited free radical scavenging activity and significantly increased non-protein thiol levels in MCF-7 cells. OCLE effectively inhibited HSV-1, HSV-2, CoxB1, and CoxB3 replication. CONCLUSIONS: The overall results showed an interesting inhibitory effect of OCLE on both MCF-7 cell survival and viral replication.


Asunto(s)
Neoplasias de la Mama , Herpesvirus Humano 1 , Orobanche , Antivirales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Herpesvirus Humano 1/fisiología , Humanos , Células MCF-7 , Extractos Vegetales/química , Extractos Vegetales/farmacología , Compuestos de Sulfhidrilo/farmacología , Replicación Viral , beta-Aminoetil Isotiourea/farmacología , beta-Aminoetil Isotiourea/uso terapéutico
4.
Int J Mol Sci ; 23(4)2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: covidwho-1715396

RESUMEN

Interferon-ß (IFN-ß) is a pleiotropic cytokine secreted in response to various pathological conditions and is clinically used for therapy of multiple sclerosis. Its application for treatment of cancer, infections and pulmonary diseases is limited by incomplete understanding of regulatory mechanisms of its functioning. Recently, we reported that IFN-ß activity is affected by interactions with S100A1, S100A4, S100A6, and S100P proteins, which are members of the S100 protein family of multifunctional Ca2+-binding proteins possessing cytokine-like activities (Int J Mol Sci. 2020;21(24):9473). Here we show that IFN-ß interacts with one more representative of the S100 protein family, the S100B protein, involved in numerous oncological and neurological diseases. The use of chemical crosslinking, intrinsic fluorescence, and surface plasmon resonance spectroscopy revealed IFN-ß binding to Ca2+-loaded dimeric and monomeric forms of the S100B protein. Calcium depletion blocks the S100B-IFN-ß interaction. S100B monomerization increases its affinity to IFN-ß by 2.7 orders of magnitude (equilibrium dissociation constant of the complex reaches 47 pM). Crystal violet assay demonstrated that combined application of IFN-ß and S100B (5-25 nM) eliminates their inhibitory effects on MCF-7 cell viability. Bioinformatics analysis showed that the direct modulation of IFN-ß activity by the S100B protein described here could be relevant to progression of multiple oncological and neurological diseases.


Asunto(s)
Interferón beta/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Animales , Células CHO , Calcio/metabolismo , Línea Celular Tumoral , Cricetulus , Humanos , Células MCF-7 , Enfermedades del Sistema Nervioso/metabolismo , Unión Proteica/fisiología
5.
Nat Commun ; 13(1): 868, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1684025

RESUMEN

SARS-CoV-2 infection is a major global public health concern with incompletely understood pathogenesis. The SARS-CoV-2 spike (S) glycoprotein comprises a highly conserved free fatty acid binding pocket (FABP) with unknown function and evolutionary selection advantage1,2. Deciphering FABP impact on COVID-19 progression is challenged by the heterogenous nature and large molecular variability of live virus. Here we create synthetic minimal virions (MiniVs) of wild-type and mutant SARS-CoV-2 with precise molecular composition and programmable complexity by bottom-up assembly. MiniV-based systematic assessment of S free fatty acid (FFA) binding reveals that FABP functions as an allosteric regulatory site enabling adaptation of SARS-CoV-2 immunogenicity to inflammation states via binding of pro-inflammatory FFAs. This is achieved by regulation of the S open-to-close equilibrium and the exposure of both, the receptor binding domain (RBD) and the SARS-CoV-2 RGD motif that is responsible for integrin co-receptor engagement. We find that the FDA-approved drugs vitamin K and dexamethasone modulate S-based cell binding in an FABP-like manner. In inflammatory FFA environments, neutralizing immunoglobulins from human convalescent COVID-19 donors lose neutralization activity. Empowered by our MiniV technology, we suggest a conserved mechanism by which SARS-CoV-2 dynamically couples its immunogenicity to the host immune response.


Asunto(s)
COVID-19/inmunología , Ácidos Grasos/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Virión/inmunología , Células A549 , Sitio Alostérico/genética , Secuencia de Aminoácidos , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Sitios de Unión/genética , COVID-19/metabolismo , COVID-19/virología , Células Cultivadas , Microscopía por Crioelectrón/métodos , Tomografía con Microscopio Electrónico/métodos , Proteínas de Unión a Ácidos Grasos/inmunología , Proteínas de Unión a Ácidos Grasos/metabolismo , Ácidos Grasos/metabolismo , Humanos , Células MCF-7 , Microscopía Confocal/métodos , Unión Proteica , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Homología de Secuencia de Aminoácido , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Virión/metabolismo , Virión/ultraestructura
6.
AAPS J ; 24(1): 8, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1555615

RESUMEN

Lipidoid nanoparticles (LNPs) are the delivery platform in Onpattro, the first FDA-approved siRNA drug. LNPs are also the carriers in the Pfizer-BioNTech and Moderna COVID-19 mRNA vaccines. While these applications have demonstrated that LNPs effectively deliver nucleic acids to hepatic and muscle cells, it is unclear if LNPs could be used for delivery of siRNA to neural cells, which are notoriously challenging delivery targets. Therefore, the purpose of this study was to determine if LNPs could efficiently deliver siRNA to neurons. Because of their potential delivery utility in either applications for the central nervous system and the peripheral nervous system, we used both cortical neurons and sensory neurons. We prepared siRNA-LNPs using C12-200, a benchmark ionizable cationic lipidoid along with helper lipids. We demonstrated using dynamic light scattering that the inclusion of both siRNA and PEG-lipid provided a stabilizing effect to the LNP particle diameters and polydispersity indices by minimizing aggregation. We found that siRNA-LNPs were safely tolerated by primary dorsal root ganglion neurons. Flow cytometry analysis revealed that Cy5 siRNA delivered via LNPs into rat primary cortical neurons showed uptake levels similar to Lipofectamine RNAiMAX-the gold standard commercial transfection agent. However, LNPs demonstrated a superior safety profile, whereas the Lipofectamine-mediated uptake was concomitant with significant toxicity. Fluorescence microscopy demonstrated a time-dependent increase in the uptake of LNP-delivered Cy5 siRNA in a human cortical neuron cell line. Overall, our results suggest that LNPs are a viable platform that can be optimized for delivery of therapeutic siRNAs to neural cells.


Asunto(s)
Ganglios Espinales/metabolismo , Lípidos/química , Nanopartículas , Neuronas/metabolismo , ARN Interferente Pequeño/administración & dosificación , Tratamiento con ARN de Interferencia , Transfección , Animales , Carbocianinas/metabolismo , Colorantes Fluorescentes/metabolismo , Ganglios Espinales/citología , Humanos , Células MCF-7 , Microscopía Fluorescente , Nanotecnología , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Factores de Tiempo
7.
Bioengineered ; 12(1): 4407-4419, 2021 12.
Artículo en Inglés | MEDLINE | ID: covidwho-1373615

RESUMEN

Widespread infection due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) has led to a global pandemic. Currently, various approaches are being taken up to develop vaccines and therapeutics to treat SARS-CoV2 infection. Consequently, the S protein has become an important target protein for developing vaccines and therapeutics against SARS-CoV2. However, the highly infective nature of SARS-CoV2 restricts experimentation with the virus to highly secure BSL3 facilities. The availability of fusion-enabled, nonreplicating, and nonbiohazardous mimics of SARS-CoV2 virus fusion, containing the viral S or S and M protein in their native conformation on mammalian cells, can serve as a useful substitute for studying viral fusion for testing various inhibitors of viral fusion. This would avoid the use of the BSL3 facility for fusion studies required to develop therapeutics. In the present study, we have developed SARS-CoV2 virus fusion mimics (SCFMs) using mammalian cells transfected with constructs coding for S or S and M protein. The fusogenic property of the mimic(s) and their interaction with the functional human ACE2 receptors was confirmed experimentally. We have also shown that such mimics can easily be used in an inhibition assay. These mimic(s) can be easily prepared on a large scale, and such SCFMs can serve as an invaluable resource for viral fusion inhibition assays and in vitro screening of antiviral agents, which can be shared/handled between labs/facilities without worrying about any biohazard while working under routine laboratory conditions, avoiding the use of BSL3 laboratory.Abbreviations :SCFM: SARS-CoV2 Virus Fusion Mimic; ACE2: Angiotensin-Converting Enzyme 2; hACE2: Human Angiotensin-Converting enzyme 2; MEF: Mouse Embryonic Fibroblasts; HBSS: Hanks Balanced Salt Solution; FBS: Fetal Bovine Serum.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Contención de Riesgos Biológicos/métodos , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Proteínas de la Matriz Viral/antagonistas & inhibidores , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Chlorocebus aethiops , Embrión de Mamíferos , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Células MCF-7 , Ratones , Imitación Molecular , Plásmidos/química , Plásmidos/metabolismo , Cultivo Primario de Células , Unión Proteica , Receptores Virales/genética , Receptores Virales/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Transfección , Células Vero , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo
8.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: covidwho-1367846

RESUMEN

Coronavirus Disease 2019 (COVID-19) remains a global health crisis, despite the development and success of vaccines in certain countries. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19, uses its spike protein to bind to the human cell surface receptor angiotensin-converting enzyme 2 (ACE2), which allows the virus to enter the human body. Using our unique cell screening technology, we identified two ACE2-binding peptoid compounds and developed dimeric derivatives (ACE2P1D1 and ACE2P2D1) that effectively blocked spike protein-ACE2 interaction, resulting in the inhibition of SARS-CoV-2 pseudovirus entry into human cells. ACE2P1D1 and ACE2P2D1 also blocked infection by a D614G mutant pseudovirus. More importantly, these compounds do not decrease ACE2 expression nor its enzyme activity (which is important in normal blood pressure regulation), suggesting safe applicability in humans.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/prevención & control , Peptidil-Dipeptidasa A/metabolismo , Peptoides/farmacología , SARS-CoV-2/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , COVID-19/virología , Humanos , Células MCF-7 , Peptoides/metabolismo , Unión Proteica/efectos de los fármacos , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Tratamiento Farmacológico de COVID-19
9.
Viruses ; 13(7)2021 06 29.
Artículo en Inglés | MEDLINE | ID: covidwho-1289028

RESUMEN

Vitis vinifera represents an important and renowned source of compounds with significant biological activity. Wines and winery bioproducts, such as grape pomace, skins, and seeds, are rich in bioactive compounds against a wide range of human pathogens, including bacteria, fungi, and viruses. However, little is known about the biological properties of vine leaves. The aim of this study was the evaluation of phenolic composition and antiviral activity of Vitis vinifera leaf extract against two human viruses: the Herpes simplex virus type 1 (HSV-1) and the pandemic and currently widespread severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). About 40 phenolic compounds were identified in the extract by HPLC-MS/MS analysis: most of them were quercetin derivatives, others included derivatives of luteolin, kaempferol, apigenin, isorhamnetin, myricetin, chrysoeriol, biochanin, isookanin, and scutellarein. Leaf extract was able to inhibit both HSV-1 and SARS-CoV-2 replication in the early stages of infection by directly blocking the proteins enriched on the viral surface, at a very low concentration of 10 µg/mL. These results are very promising and highlight how natural extracts could be used in the design of antiviral drugs and the development of future vaccines.


Asunto(s)
Antivirales/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Extractos Vegetales/farmacología , Hojas de la Planta/química , SARS-CoV-2/efectos de los fármacos , Vitis/química , Células A549 , Animales , Productos Biológicos/análisis , Productos Biológicos/farmacología , Línea Celular , Chlorocebus aethiops , Cromatografía Líquida de Alta Presión , Humanos , Células MCF-7 , Fenoles/farmacología , Extractos Vegetales/análisis , Espectrometría de Masas en Tándem , Células Vero
10.
Int J Biol Macromol ; 184: 297-312, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1265684

RESUMEN

COVID-19 caused by SARS-CoV-2 corona virus has become a global pandemic. In the absence of drugs and vaccine, and premises of time, efforts and cost required for their development, natural resources such as herbs are anticipated to provide some help and may also offer a promising resource for drug development. Here, we have investigated the therapeutic prospective of Ashwagandha for the COVID-19 pandemic. Nine withanolides were tested in silico for their potential to target and inhibit (i) cell surface receptor protein (TMPRSS2) that is required for entry of virus to host cells and (ii) viral protein (the main protease Mpro) that is essential for virus replication. We report that the withanolides possess capacity to inhibit the activity of TMPRSS2 and Mpro. Furthermore, withanolide-treated cells showed downregulation of TMPRSS2 expression and inhibition of SARS-CoV-2 replication in vitro, suggesting that Ashwagandha may provide a useful resource for COVID-19 treatment.


Asunto(s)
Antivirales/farmacología , Extractos Vegetales/química , SARS-CoV-2/fisiología , Serina Endopeptidasas/metabolismo , Proteínas de la Matriz Viral/metabolismo , Witanólidos/farmacología , Células A549 , Antivirales/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Regulación hacia Abajo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Modelos Moleculares , Simulación de Dinámica Molecular , Conformación Proteica , SARS-CoV-2/efectos de los fármacos , Serina Endopeptidasas/química , Proteínas de la Matriz Viral/química , Internalización del Virus/efectos de los fármacos , Witanólidos/química
11.
J Extracell Vesicles ; 10(8): e12110, 2021 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1258076

RESUMEN

Circulating nucleic acids, encapsulated within small extracellular vesicles (EVs), provide a remote cellular snapshot of biomarkers derived from diseased tissues, however selective isolation is critical. Current laboratory-based purification techniques rely on the physical properties of small-EVs rather than their inherited cellular fingerprints. We established a highly-selective purification assay, termed EV-CATCHER, initially designed for high-throughput analysis of low-abundance small-RNA cargos by next-generation sequencing. We demonstrated its selectivity by specifically isolating and sequencing small-RNAs from mouse small-EVs spiked into human plasma. Western blotting, nanoparticle tracking, and transmission electron microscopy were used to validate and quantify the capture and release of intact small-EVs. As proof-of-principle for sensitive detection of circulating miRNAs, we compared small-RNA sequencing data from a subset of small-EVs serum-purified with EV-CATCHER to data from whole serum, using samples from a small cohort of recently hospitalized Covid-19 patients. We identified and validated, only in small-EVs, hsa-miR-146a and hsa-miR-126-3p to be significantly downregulated with disease severity. Separately, using convalescent sera from recovered Covid-19 patients with high anti-spike IgG titers, we confirmed the neutralizing properties, against SARS-CoV-2 in vitro, of a subset of small-EVs serum-purified by EV-CATCHER, as initially observed with ultracentrifuged small-EVs. Altogether our data highlight the sensitivity and versatility of EV-CATCHER.


Asunto(s)
Vesículas Extracelulares/química , Técnicas Inmunológicas/métodos , Animales , Secreciones Corporales/química , COVID-19/sangre , COVID-19/fisiopatología , Chlorocebus aethiops , MicroARN Circulante , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Células MCF-7 , Ratones , Células RAW 264.7 , Índice de Severidad de la Enfermedad , Células Vero
12.
ACS Nano ; 15(1): 1167-1178, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: covidwho-1014984

RESUMEN

Existing methods for RNA diagnostics, such as reverse transcription PCR (RT-PCR), mainly rely on nucleic acid amplification (NAA) and RT processes, which are known to introduce substantial issues, including amplification bias, cross-contamination, and sample loss. To address these problems, we introduce a confinement effect-inspired Cas13a assay for single-molecule RNA diagnostics, eliminating the need for NAA and RT. This assay involves confining the RNA-triggered Cas13a catalysis system in cell-like-sized reactors to enhance local concentrations of target and reporter simultaneously, via droplet microfluidics. It achieves >10 000-fold enhancement in sensitivity when compared to the bulk Cas13a assay and enables absolute digital single-molecule RNA quantitation. We experimentally demonstrate its broad applicability for precisely counting microRNAs, 16S rRNAs, and SARS-CoV-2 RNA from synthetic sequences to clinical samples with excellent accuracy. Notably, this direct RNA diagnostic technology enables detecting a wide range of RNA molecules at the single-molecule level. Moreover, its simplicity, universality, and excellent quantification capability might render it to be a dominant rival to RT-qPCR.


Asunto(s)
Sistemas CRISPR-Cas , Microfluídica , ARN/análisis , Línea Celular Tumoral , Enterococcus faecalis , Escherichia coli , Humanos , Klebsiella pneumoniae , Células MCF-7 , MicroARNs/análisis , Pseudomonas aeruginosa , ARN Ribosómico 16S/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , SARS-CoV-2/genética , Staphylococcus aureus
13.
Arch Med Res ; 52(1): 48-57, 2021 01.
Artículo en Inglés | MEDLINE | ID: covidwho-893598

RESUMEN

BACKGROUND: Ras-GTPase activating protein SH3-domain-binding proteins (G3BP) are a small family of RNA-binding proteins implicated in regulating gene expression. Changes in expression of G3BPs are correlated to several cancers including thyroid, colon, pancreatic and breast cancer. G3BPs are important regulators of stress granule (SG) formation and function. SG are ribonucleoprotein (RNP) particles that respond to cellular stresses to triage mRNA resulting in transcripts being selectively degraded, stored or translated resulting in a change of gene expression which confers a survival response to the cell. These changes in gene expression contribute to the development of drug resistance. Many RNA viruses, including Chikungunya (and potentially Coronavirus), dismantle SG so that the cell cannot respond to the viral infection. Non-structural protein 3 (nsP3), from the Chikungunya virus, has been shown to translocate G3BP away from SG. Interestingly in cancer cells, the formation of SG is correlated to drug-resistance and blocking SG formation has been shown to reestablish the efficacy of the anticancer drug bortezomib. METHODS: Chikungunya nsP3 was transfected into breast cancer cell lines T47D and MCF7 to disrupt SG formation. Changes in the cytotoxicity of bortezomib were measured. RESULTS: Bortezomib cytotoxicity in breast cancer cell lines changed with a 22 fold decrease in its IC50 for T47D and a 7 fold decrease for MCF7 cells. CONCLUSIONS: Chikungunya nsP3 disrupts SG formation. As a result, it increases the cytotoxicity of the FDA approved drug, bortezomib. In addition, the increased cytotoxicity appears to correlate to improved bortezomib selectivity when compared to control cell lines.


Asunto(s)
Bortezomib/farmacología , Fiebre Chikungunya/tratamiento farmacológico , Virus Chikungunya/genética , Gránulos Citoplasmáticos/metabolismo , ADN Helicasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Fiebre Chikungunya/metabolismo , Fiebre Chikungunya/patología , Virus Chikungunya/metabolismo , Chlorocebus aethiops , Gránulos Citoplasmáticos/efectos de los fármacos , Gránulos Citoplasmáticos/patología , ADN Helicasas/genética , Regulación hacia Abajo , Resistencia a Antineoplásicos , Femenino , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Proteínas de Unión a Poli-ADP-Ribosa/genética , ARN Helicasas/genética , Proteínas con Motivos de Reconocimiento de ARN/genética , Transfección , Células Vero , Proteínas no Estructurales Virales/administración & dosificación , Proteínas no Estructurales Virales/genética
14.
J Cell Mol Med ; 24(21): 12864-12868, 2020 11.
Artículo en Inglés | MEDLINE | ID: covidwho-780917

RESUMEN

The SARS-coronavirus 2 is the aetiologic agent COVID-19. ACE2 has been identified as a cell entry receptor for the virus. Therefore, trying to understand how the gene is controlled has become a major goal. We silenced the expression of STAT3α and STAT3ß, and found that while silencing STAT3α causes an increase in ACE2 expression, silencing STAT3ß causes the opposite effect. Studying the role of STAT3 in ACE2 expression will shed light on the molecular events that contribute to the progression of the disease and that the different roles of STAT3α and STAT3ß in that context must be taken in consideration. Our results place STAT3 in line with additional potential therapeutic targets for treating COVID-19 patients.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Factor de Transcripción STAT3/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Sitios de Unión , COVID-19 , Humanos , Células MCF-7 , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , SARS-CoV-2/efectos de los fármacos , Factor de Transcripción STAT3/genética
15.
Mol Syst Biol ; 16(7): e9628, 2020 07.
Artículo en Inglés | MEDLINE | ID: covidwho-707164

RESUMEN

The COVID-19 pandemic caused by SARS-CoV-2 has is a global health challenge. Angiotensin-converting enzyme 2 (ACE2) is the host receptor for SARS-CoV-2 entry. Recent studies have suggested that patients with hypertension and diabetes treated with ACE inhibitors (ACEIs) or angiotensin receptor blockers have a higher risk of COVID-19 infection as these drugs could upregulate ACE2, motivating the study of ACE2 modulation by drugs in current clinical use. Here, we mined published datasets to determine the effects of hundreds of clinically approved drugs on ACE2 expression. We find that ACEIs are enriched for ACE2-upregulating drugs, while antineoplastic agents are enriched for ACE2-downregulating drugs. Vorinostat and isotretinoin are the top ACE2 up/downregulators, respectively, in cell lines. Dexamethasone, a corticosteroid used in treating severe acute respiratory syndrome and COVID-19, significantly upregulates ACE2 both in vitro and in vivo. Further top ACE2 regulators in vivo or in primary cells include erlotinib and bleomycin in the lung and vancomycin, cisplatin, and probenecid in the kidney. Our study provides leads for future work studying ACE2 expression modulators.


Asunto(s)
Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Células A549 , Enzima Convertidora de Angiotensina 2 , Betacoronavirus , Bleomicina/farmacología , COVID-19 , Dexametasona/farmacología , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Clorhidrato de Erlotinib/farmacología , Flufenazina/farmacología , Células HEK293 , Humanos , Riñón/efectos de los fármacos , Pulmón/efectos de los fármacos , Células MCF-7 , Pandemias , Peptidil-Dipeptidasa A , SARS-CoV-2 , Biología de Sistemas , Regulación hacia Arriba , Vemurafenib/farmacología , Tratamiento Farmacológico de COVID-19
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA