Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Biochem J ; 475(6): 1059-1062, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29559581

RESUMEN

The HIV-1 tat gene encodes a small 86-104 amino acid protein depending on the HIV-1 strain. Tat is essential for HIV-1 replication through interactions with numerous cellular transcription factors. The interaction between Tat and P-TEFb, which is a cellular protein complex composed of cyclin T1 and CDK9, delivers P-TEFb to the newly transcribed viral mRNAs where phosphorylation of RNA polymerase II by CDK9 leads to highly efficient mRNA transcription. It has long been recognized that Tat is a potential anti-HIV-1 target and possibly a viral Achilles' heel. However, specifically targeting Tat without affecting normal host cell functions has been challenging. Means to inactivate Tat have been reported that includes small compounds, transdominant negative Tat proteins, and by plant-derived antivirals. Investigations of these agents have reported encouraging outcomes that inform and may hopefully affect strategies for a functional HIV-1 cure.


Asunto(s)
VIH-1/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Ciclina T , Quinasa 9 Dependiente de la Ciclina/genética , Glicósido Hidrolasas , ARN , Transcripción Genética , Activación Transcripcional
2.
Virol J ; 15(1): 182, 2018 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-30477508

RESUMEN

Cellular protein eukaryotic translation elongation factor 1A (eEF1A) is an actin binding protein that plays a role in the formation of filamentous actin (F-actin) bundles. F-Actin regulates multiple stages of respiratory syncytial virus (RSV) replication including assembly and budding. Our previous study demonstrated that eEF1A knock-down significantly reduced RSV replication. Here we investigated if the eEF1A function in actin bundle formation was important for RSV replication and release. To investigate this, eEF1A function was impaired in HEp-2 cells by either knock-down of eEF1A with siRNA, or treatment with an eEF1A inhibitor, didemnin B (Did B). Cell staining and confocal microscopy analysis showed that both eEF1A knock-down and treatment with Did B resulted in disruption of cellular stress fiber formation and elevated accumulation of F-actin near the plasma membrane. When treated cells were then infected with RSV, there was also reduced formation of virus-induced cellular filopodia. Did B treatment, similarly to eEF1A knock-down, reduced the release of infectious RSV, but unlike eEF1A knock-down, did not significantly affect RSV genome replication. The lower infectious virus production in Did B treated cells also reduced RSV-induced cell death. In conclusion, the cellular factor eEF1A plays an important role in the regulation of F-actin stress fiber formation required for RSV assembly and release.


Asunto(s)
Actinas/metabolismo , Factor 1 de Elongación Peptídica/genética , Virus Sincitial Respiratorio Humano/fisiología , Fibras de Estrés/fisiología , Replicación Viral , Actinas/genética , Línea Celular Tumoral , Depsipéptidos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Técnicas de Silenciamiento del Gen , Humanos , Seudópodos/fisiología , Seudópodos/virología , Virus Sincitial Respiratorio Humano/genética
3.
Biochim Biophys Acta ; 1863(2): 254-62, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26611710

RESUMEN

The human immunodeficiency virus (HIV)-1 transactivator protein Tat is known to play a key role in HIV infection, integrally related to its role in the host cell nucleus/nucleolus. Here we show for the first time that Tat localisation can be modulated by specific methylation, whereby overexpression of active but not catalytically inactive PRMT6 methyltransferase specifically leads to exclusion of Tat from the nucleolus. An R52/53A mutated Tat derivative does not show this redistribution, implying that R52/53, within Tat's nuclear/nucleolar localisation signal, are the targets of PRMT6 activity. Analysis using fluorescence recovery after photobleaching indicate that Tat nucleolar accumulation is largely through binding to nucleolar components, with methylation of Tat by PRMT6 preventing this. To our knowledge, this is the first report of specific protein methylation inhibiting nucleolar retention.


Asunto(s)
Nucléolo Celular/metabolismo , VIH-1/metabolismo , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Arginina/genética , Arginina/metabolismo , Células COS , Chlorocebus aethiops , Electroforesis en Gel de Poliacrilamida , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , VIH-1/genética , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Metilación , Microscopía Confocal , Mutación , Señales de Localización Nuclear/genética , Proteínas Nucleares/genética , Unión Proteica , Proteína-Arginina N-Metiltransferasas/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Proteína Fluorescente Roja
4.
PLoS Pathog ; 11(12): e1005289, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26624286

RESUMEN

Reverse transcription is the central defining feature of HIV-1 replication. We previously reported that the cellular eukaryotic elongation factor 1 (eEF1) complex associates with the HIV-1 reverse transcription complex (RTC) and the association is important for late steps of reverse transcription. Here we show that association between the eEF1 and RTC complexes occurs by a strong and direct interaction between the subunit eEF1A and reverse transcriptase (RT). Using biolayer interferometry and co-immunoprecipitation (co-IP) assays, we show that association between the eEF1 and RTC complexes occurs by a strong (KD ~3-4 nM) and direct interaction between eEF1A and reverse transcriptase (RT). Biolayer interferometry analysis of cell lysates with titrated levels of eEF1A indicates it is a predominant cellular RT binding protein. Both the RT thumb and connection domains are required for interaction with eEF1A. A single amino acid mutation, W252A, within the thumb domain impaired co-IP between eEF1A and RT, and also significantly reduced the efficiency of late reverse transcription and virus replication when incorporated into infectious HIV-1. Molecular modeling analysis indicated that interaction between W252 and L303 are important for RT structure, and their mutation to alanine did not impair heterodimerisation, but negatively impacted interaction with eEF1A. Didemnin B, which specifically binds eEF1A, potently inhibited HIV-1 reverse transcription by greater than 2 logs at subnanomolar concentrations, especially affecting reverse transcription late DNA synthesis. Analysis showed reduced levels of RTCs from HIV-1-infected HEK293T treated with didemnin B compared to untreated cells. Interestingly, HIV-1 with a W252A RT mutation was resistant to didemnin B negative effects showing that didemnin B affects HIV-1 by targeting the RT-eEF1A interaction. The combined evidence indicates a direct interaction between eEF1A and RT is crucial for HIV reverse transcription and replication, and the RT-eEF1A interaction is a potential drug target.


Asunto(s)
Infecciones por VIH/metabolismo , Transcriptasa Inversa del VIH/metabolismo , VIH-1/fisiología , Factor 1 de Elongación Peptídica/metabolismo , Transcripción Reversa/fisiología , Replicación Viral/fisiología , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Inmunoprecipitación
5.
Virol J ; 14(1): 52, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28288662

RESUMEN

BACKGROUND: Nullbasic is a mutant HIV-1 Tat protein that inhibits HIV-1 replication via three independent mechanisms that disrupts 1) reverse transcription of the viral RNA genome into a DNA copy, 2) HIV-1 Rev protein function required for viral mRNA transport from the nucleus to the cytoplasm and 3) HIV-1 mRNA transcription by RNA Polymerase II. The Nullbasic protein is derived from the subtype B strain HIV-1BH10 and has only been tested against other HIV-1 subtype B strains. However, subtype B strains only account for ~10% of HIV-1 infections globally and HIV-1 Tat sequences vary between subtypes especially for subtype C, which is responsible for ~50% HIV-1 infection worldwide. These differences could influence the ability of Tat to interact with RNA and cellular proteins and thus could affect the antiviral activity of Nullbasic. Therefore, Nullbasic was tested against representative HIV-1 strains from subtypes C, D and A/D recombinant to determine if it can inhibit their replication. METHODS: Nullbasic was delivered to human cells using a self-inactivating (SIN) γ-retroviral system. We evaluated Nullbasic-mCherry (NB-mCh) fusion protein activity against the HIV-1 strains in TZM-bl cell lines for inhibition of transactivation and virus replication. We also examined antiviral activity of Nullbasic-ZsGreen1 (NB-ZSG1) fusion protein against the same strains in primary CD4+ T cells. The Nullbasic expression was monitored by western blot and flow cytometry. The effects of Nullbasic on primary CD4+ T cells cytotoxicity, proliferation and apoptosis were also examined. RESULTS: The results show that Nullbasic inhibits Tat-mediated transactivation and virus replication of all the HIV-1 strains tested in TZM-bl cells. Importantly, Nullbasic inhibits replication of the HIV-1 strains in primary CD4+ T cells without affecting cell proliferation, cytotoxicity or level of apoptotic cells. CONCLUSION: A SIN-based γ-retroviral vector used to express Nullbasic fusion proteins improved protein expression particularly in primary CD4+ T cells. Nullbasic has antiviral activity against all strains from the subtypes tested although small differences in viral inhibition were observed. Further improvement of in γ-retroviral vector stable expression of Nullbasic expression may have utility in a future gene therapy approach applicable to genetically diverse HIV-1 strains.


Asunto(s)
Antivirales/metabolismo , Genotipo , VIH-1/fisiología , Proteínas Mutantes/metabolismo , Replicación Viral , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD4-Positivos/virología , Células Cultivadas , VIH-1/clasificación , VIH-1/genética , Humanos , Proteínas Mutantes/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
6.
J Virol ; 89(9): 4827-36, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25673710

RESUMEN

UNLABELLED: Previously, we reported that a mutant of Tat referred to as Nullbasic inhibits HIV-1 reverse transcription although the mechanism of action is unknown. Here we show that Nullbasic is a reverse transcriptase (RT) binding protein that targets the reverse transcription complex rather than directly inhibiting RT activity. An interaction between Nullbasic and RT was observed by using coimmunoprecipitation and pulldown assays, and a direct interaction was measured by using a biolayer interferometry assay. Mixtures of recombinant 6×His-RT and Nullbasic-FLAG-V5-6×His at molar ratios of up to 1:20,000 did not inhibit RT activity in standard homopolymer primer template assays. An analysis of virus made by cells that coexpressed Nullbasic showed that Nullbasic copurified with virus particles, indicating that it was a virion protein. In addition, analysis of reverse transcription complexes (RTCs) isolated from cells infected with wild type or Nullbasic-treated HIV-1 showed that Nullbasic reduced the levels of viral DNA in RTC fractions. In addition, a shift in the distribution of viral DNA and CAp24 to less-dense non-RTC fractions was observed, indicating that RTC activity from Nullbasic-treated virus was impaired. Further analysis showed that viral cores isolated from Nullbasic-treated HIV undergo increased disassembly in vitro compared to untreated HIV-1. To our knowledge, this is the first description of an antiviral protein that inhibits reverse transcription by targeting the RTC and affecting core stability. IMPORTANCE: HIV-1 infection is treated by using combinations of antiretroviral drugs that target independent steps of virus replication. A newly described antiviral protein called Nullbasic can also inhibit a combination of different steps in virus replication (transcription, reverse transcription, and Rev-mediated viral mRNA transport), although the precise mechanism of action is unknown. This study shows that Nullbasic can inhibit reverse transcription by binding to the viral enzyme called reverse transcriptase, which results in accelerated uncoating of the viral core and instability of the viral apparatus called the reverse transcription complex (RTC). This unique antiviral activity may inform development of other RTC inhibitors, as well as providing a unique investigative tool for dissecting the RTC cellular composition.


Asunto(s)
VIH-1/fisiología , Transcripción Reversa , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Centrifugación , Transcriptasa Inversa del VIH/antagonistas & inhibidores , Transcriptasa Inversa del VIH/metabolismo , VIH-1/genética , Inmunoprecipitación , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Unión Proteica , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
7.
Virol J ; 12: 118, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-26242867

RESUMEN

BACKGROUND: The cellular protein eukaryotic translation elongation factor 1A (eEF1A) binds to aminoacylated transfer RNAs and delivers them to the ribosome during translation. eEF1A also binds to RNA secondary structures present in genomes of several RNA viruses and plays important roles in their replication. As a RNA binding protein, whether eEF1A can bind with HIV-1 genomic RNA has not been investigated and was the aim of the study. METHODS: RNA-protein interaction was determined by reversible crosslink co-immunoprecipitation (RC-Co-IP) and biolayer Interferometry assay (BLI). eEF1A binding region within RNA was mapped by deletion and mutation analysis. Virus with genomic RNA mutations were examined for eEF1A-RT interaction by proximity ligation assay, for reverse transcription by qPCR and for replication by CAp24 ELISA in cells. RESULTS: The interaction of eEF1A with 5'UTR of HIV-1 genomic RNA was detected in cells and in vitro. Truncation and substitution mutations in the 5'UTR RNA demonstrated that a stem-loop formed by nucleotides 142 to 170, which encompass a reported tRNA anticodon-like-element, binds to eEF1A. Mutations that altered the stem-loop structure by changing two highly conserved sequence clusters in the stem-loop region result in reduction of the interaction with eEF1A in vitro. HIV-1 virus harbouring the same 5'UTR mutations significantly reduced the interaction of eEF1A with HIV-1 reverse transcription complex (RTC), reverse transcription and replication. CONCLUSION: eEF1A interacts with 5'UTR of HIV-1 genomic RNA and the interaction is important for late DNA synthesis in reverse transcription.


Asunto(s)
Regiones no Traducidas 5' , Genoma Viral , VIH-1/genética , Factor 1 de Elongación Peptídica/metabolismo , ARN Viral , Transcripción Reversa , Línea Celular , Humanos , Secuencias Invertidas Repetidas , Mutación , Conformación de Ácido Nucleico , Unión Proteica , ARN Viral/química , ARN Viral/genética
8.
Retrovirology ; 11: 121, 2014 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-25496916

RESUMEN

BACKGROUND: Previously we described a transdominant negative mutant of the HIV-1 Tat protein, termed Nullbasic, that downregulated the steady state levels of unspliced and singly spliced viral mRNA, an activity caused by inhibition of HIV-1 Rev activity. Nullbasic also altered the subcellular localizations of Rev and other cellular proteins, including CRM1, B23 and C23 in a Rev-dependent manner, suggesting that Nullbasic may disrupt Rev function and trafficking by intervening with an unidentified component of the Rev nucleocytoplasmic transport complex. RESULTS: To seek a possible mechanism that could explain how Nullbasic inhibits Rev activity, we used a proteomics approach to identify host cellular proteins that interact with Nullbasic. Forty-six Nullbasic-binding proteins were identified by mass spectrometry including the DEAD-box RNA helicase, DDX1. To determine the effect of DDX1 on Nullbasic-mediated Rev activity, we performed cell-based immunoprecipitation assays, Rev reporter assays and bio-layer interferometry (BLI) assays. Interaction between DDX1 and Nullbasic was observed by co-immunoprecipitation of Nullbasic with endogenous DDX1 from cell lysates. BLI assays showed a direct interaction between Nullbasic and DDX1. Nullbasic affected DDX1 subcellular distribution in a Rev-independent manner. Interestingly overexpression of DDX1 in cells not only restored Rev-dependent mRNA export and gene expression in a Rev reporter assay but also partly reversed Nullbasic-induced Rev subcellular mislocalization. Moreover, HIV-1 wild type Tat co-immunoprecipitated with DDX1 and overexpression of Tat could rescue the unspliced viral mRNA levels inhibited by Nullbasic in HIV-1 expressing cells. CONCLUSIONS: Nullbasic was used to further define the complex mechanisms involved in the Rev-dependent nuclear export of the 9 kb and 4 kb viral RNAs. All together, these data indicate that DDX1 can be sequestered by Nullbasic leading to destabilization of the Rev nucleocytoplasmic transport complex and decreased levels of Rev-dependent viral transcripts. The outcomes support a role for DDX1 in maintenance of a Rev nuclear complex that transports viral RRE-containing mRNA to the cytoplasm. To our knowledge Nullbasic is the first anti-HIV protein that specifically targets the cellular protein DDX1 to block Rev's activity. Furthermore, our research raises the possibility that wild type Tat may play a previously unrecognized but very important role in Rev function.


Asunto(s)
ARN Helicasas DEAD-box/antagonistas & inhibidores , VIH-1/fisiología , Replicación Viral , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Línea Celular , VIH-1/genética , Humanos , Inmunoprecipitación , Espectrometría de Masas , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mapeo de Interacción de Proteínas , Proteómica , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
9.
Virol J ; 10: 207, 2013 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-23800116

RESUMEN

BACKGROUND: Protein arginine methyltransferase 6 (PRMT6) can methylate the HIV-1 Tat, Rev and nucleocapsid proteins in a manner that diminishes each of their functions in in vitro assays, and increases the stability of Tat in human cells. In this study, we explored the relationship between PRMT6 and HIV-1 Tat by determining the domains in each protein required for interaction. METHODS: Through domain mapping and immunoprecipitation experiments, we determined that both the amino and carboxyl termini of PRMT6, and the activation domain within Tat are essential for interaction. Mutation of the basic domain of Tat did not affect the ability of PRMT6 to interact with Tat. RESULTS: We next used the A549 human alveolar adenocarcinoma cell line, which naturally expresses undetectable levels of PRMT6, as a model for testing the effects of PRMT6 on Tat stability, transactivation, and HIV-1 replication. As previously observed, steady state levels and the protein half-life of Tat were increased by the ectopic expression of PRMT6. However, no down regulation of Tat transactivation function was observed, even with over 300-fold molar excess of PRMT6 plasmid. We also observed no negative effect on HIV-1 infectivity when A549 producer cells overexpressed PRMT6. CONCLUSIONS: We show that PRMT6 requires the activation domain, but surprisingly not the basic domain, of Tat for protein interaction. This interaction between Tat and PRMT6 may impact upon pathogenic effects attributed to Tat during HIV-1 infection other than its function during transactivation.


Asunto(s)
VIH-1/fisiología , Interacciones Huésped-Patógeno , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Activación Transcripcional , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Línea Celular , Células Epiteliales/virología , Humanos , Inmunoprecipitación , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína
10.
Front Immunol ; 13: 972503, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36211350

RESUMEN

Background: Whether intensity-modulated radiotherapy (IMRT) can enhance the efficacy of the programmed death (PD)-1 inhibitors combined with anti-angiogenic therapy for hepatocellular carcinoma (HCC) is unclear. Therefore, we conducted this multicenter retrospective study to investigate the efficacy of the combination of PD-1 inhibitors with anti-angiogenic therapy and IMRT. Methods: From April 2019 to March 2022, a total of 197 patients with HCC [combination of PD-1 inhibitors with anti-angiogenic therapy and IMRT (triple therapy group), 54; PD-1 inhibitors plus anti-angiogenic therapy (control group), 143] were included in our study. Propensity score matching (PSM) was applied to identify two groups with similar baselines. The objective response rate (ORR), overall survival (OS), and progression-free survival (PFS) of the two groups were compared before and after matching. Results: Prior to PSM, the triple therapy group had higher ORR (42.6% vs 24.5%, P = 0.013) and more superior median OS (mOS) (20.1 vs 13.3 months, P = 0.009) and median PFS (mPFS) (8.7 vs 5.4 months, P = 0.001) than the control group. Following PSM, the triple therapy group still exhibited better mPFS (8.7 vs 5.4 months, P = 0.013) and mOS (18.5 vs 12.6 months, P = 0.043) than the control group. However, the ORR of the two groups was similar (40% vs 25%, P = 0.152). No significant difference was observed in the treatment-related adverse events between the two groups (P < 0.05 for all). Conclusions: The combination of PD-1 inhibitors with anti-angiogenic therapy and IMRT for HCC is a promising regimen.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Radioterapia de Intensidad Modulada , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/radioterapia , Humanos , Inhibidores de Puntos de Control Inmunológico , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/radioterapia , Puntaje de Propensión , Radioterapia de Intensidad Modulada/efectos adversos , Estudios Retrospectivos
11.
Front Oncol ; 12: 873830, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35982979

RESUMEN

Aim: A programmed death 1 (PD-1) inhibitor coupled with radiotherapy and antiangiogenic therapy is a potential therapeutic strategy for advanced hepatocellular carcinoma (HCC). We aimed to determine if circulating tumor cells (CTCs) positive for programmed death-ligand 1 (PD-L1) could be employed as a predictive biomarker in HCC patients receiving triple therapy. Methods: In this study, HCC patients received a PD-1 inhibitor in combination with intensity-modulated radiotherapy (IMRT) and antiangiogenic therapy. Following IMRT, the PD-1 inhibitor was administrated once every 3 weeks, while the antiangiogenic drug was given once a day. Treatment was continued until the disease progressed. Two mL of peripheral blood was collected at baseline, 1 month, and 3 months after treatment for CTC enrichment using the CytoSorter® system with a CytoSorter™ CTC PD-L1 Kit (Watson Biotech., China). Result: A total of 47 HCC patients receiving the triple therapy were enrolled in this study. Patients with < 2 PD-L1+ CTCs at baseline had a higher objective response rate (ORR) and longer overall survival (OS) than those with ≥ 2 PD-L1+ CTCs (56.5% vs. 16.7%, p = 0.007; not reach vs. 10.8 months, p = 0.001, respectively). The count of PD-L1+ CTCs was found to be an independent predictive biomarker of OS. Furthermore, the objective response was more likely to be achieved in patients with a dynamic decrease in PD-L1+ CTC counts at 1 month after treatment. Conclusions: Our study demonstrated that PD-L1+ CTCs could be a predictive biomarker for HCC patients receiving PD-1 inhibitors in combination with IMRT and antiangiogenic therapy.

12.
Hepatol Int ; 16(4): 858-867, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35729469

RESUMEN

BACKGROUND: The optimal locoregional treatment for hepatocellular carcinoma (HCC) patients with portal vein tumor thrombus (PVTT) is unclear. This study aimed to investigate the efficacy of Gamma knife radiosurgery (GKR) versus transcatheter arterial chemoembolization (TACE) in HCC patients with PVTT. METHODS: This retrospective study included 544 HCC patients with PVTT (GKR, 202; TACE, 342). Propensity score matching (PSM) analysis identified 171 matched pairs of patients. The primary endpoint was overall survival (OS). RESULTS: Before PSM, the GKR group exhibited longer median OS (mOS) than the TACE group (17.2 vs. 8.0 months, p < 0.001). We followed the Cheng's classification for PVTT. In the subgroup analysis, GKR was associated with significantly longer mOS for patients with PVTT II-IV (17.5 vs. 8.7 months, p < 0.001; 17.2 vs. 7.8 months, p = 0.001; 14.5 vs. 6.5 months, p = 0.001, respectively) and comparable OS for patients with PVTT I. After PSM, the GKR group had also a longer mOS than the TACE group (15.8 vs. 10.4 months, p < 0.001). In the subgroup analysis, the GKR group demonstrated superior mOS for patients with PVTT II-IV (all p < 0.05) and comparable OS for patients with PVTT I. CONCLUSIONS: GKR was associated better OS than TACE in HCC patients with PVTT, especially for patients with PVTT II-IV. CLINICAL TRIALS REGISTRATION: The study was registered in the Chinese Clinical Trials Registry under the registration number ChiCTR2100051057.


Asunto(s)
Carcinoma Hepatocelular , Quimioembolización Terapéutica , Neoplasias Hepáticas , Radiocirugia , Trombosis , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Humanos , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Vena Porta/patología , Puntaje de Propensión , Estudios Retrospectivos , Trombosis/cirugía , Resultado del Tratamiento
13.
Hepatol Int ; 16(5): 1208-1219, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35972640

RESUMEN

BACKGROUND: Although the diagnostic value of plasma heat-shock protein 90α (HSP90α) in hepatocellular carcinoma (HCC) has been previously reported, the causal effect of the plasma HSP90α levels on HCC prognosis remains largely unclear. To this extent, we sought to assess whether the plasma HSP90α acts as a prognostic factor for HCC patients. METHODS: A total of 2150 HCC patients were included in this retrospective study between August 2016 and July 2021. Plasma HSP90α levels were tested within a week before treatment and their association with prognosis was assessed. RESULTS: An optimal cutoff value of 143.5 for the HSP90α based on the overall survival (OS) was determined using the X-tile software. HCC patients with HSP90α < 143.5 ng/mL (low HSP90α) before and after propensity score matching (PSM) indicated longer median OS (mOS) relative to those with HSP90α ≥ 143.5 ng/mL (high HSP90α) (37.0 vs. 9.0 months, p < 0.001; 19.2 vs. 9.6 months, p < 0.001; respectively). In addition, the high HSP90α plasma level is an independent poor prognostic factor for OS in HCC patients. In our subgroup analysis, including the supportive care group, surgery group, transarterial chemoembolization (TACE) group, adjuvant TACE group, an immune checkpoint inhibitor (ICI) plus targeted therapy group, and TACE plus ICI group, the high HSP90α group demonstrated better OS compared to the low HSP90α group. Moreover, in the supportive care, TACE, ICI plus targeted therapy, TACE plus ICI groups, and high HSP90α levels were also an independent poor prognostic factors for OS. CONCLUSIONS: Our study confirmed that the plasma HSP90α level can be used as a prognostic biomarker for HCC.


Asunto(s)
Carcinoma Hepatocelular , Quimioembolización Terapéutica , Neoplasias Hepáticas , Biomarcadores , Carcinoma Hepatocelular/patología , Proteínas HSP90 de Choque Térmico , Proteínas de Choque Térmico , Humanos , Inhibidores de Puntos de Control Inmunológico , Neoplasias Hepáticas/patología , Pronóstico , Estudios Retrospectivos
14.
Commun Biol ; 4(1): 557, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976375

RESUMEN

Dengue virus (DENV) is spread from human to human through the bite of the female Aedes aegypti mosquito and leads to about 100 million clinical infections yearly. Treatment options and vaccine availability for DENV are limited. Defective interfering particles (DIPs) are considered a promising antiviral approach but infectious virus contamination has limited their development. Here, a DENV-derived DIP production cell line was developed that continuously produced DENV-free DIPs. The DIPs contained and could deliver to cells a DENV serotype 2 subgenomic defective-interfering RNA, which was originally discovered in DENV infected patients. The DIPs released into cell culture supernatant were purified and could potently inhibit replication of all DENV serotypes in cells. Antiviral therapeutics are limited for many viral infection. The DIP system described could be re-purposed to make antiviral DIPs for many other RNA viruses such as SARS-CoV-2, yellow fever, West Nile and Zika viruses.


Asunto(s)
Virus Defectuosos , Vacunas contra el Dengue/uso terapéutico , Virus del Dengue/crecimiento & desarrollo , Dengue/prevención & control , Replicación Viral , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Virus Defectuosos/genética , Virus Defectuosos/metabolismo , Dengue/virología , Virus del Dengue/genética , Virus del Dengue/metabolismo , Genes Reporteros , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , ARN Viral/biosíntesis , ARN Viral/genética , Células Vero , Carga Viral
15.
Front Immunol ; 12: 604974, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679742

RESUMEN

Antigen (Ag)-mediated mast cell activation plays a critical role in the immunopathology of IgE-dependent allergic diseases. Restraining the signaling cascade that regulates the release of mast cell-derived inflammatory mediators is an attractive therapeutic strategy to treat allergic diseases. Orosomucoid-like-3 (ORMDL3) regulates the endoplasmic reticulum stress (ERS)-induced unfolded protein response (UPR) and autophagy. Although ERS/UPR/autophagy pathway is crucial in Ag-induced mast cell activation, it is unknown whether ORMDL3 regulates the ERS/UPR/autophagy pathway during mast cell activation. In this study, we found that ORMDL3 expression was downregulated in Ag-activated MC/9 cells. Overexpression of ORMDL3 significantly inhibited degranulation, and cytokine/chemokine production, while the opposite effect was observed with ORMDL3 knockdown in MC/9 cells. Importantly, ORMDL3 overexpression upregulated mediators of ERS-UPR (SERCA2b, ATF6) and autophagy (Beclin 1 and LC3BII). Knockdown of ATF6 and/or inhibition of autophagy reversed the decreased degranulation and cytokine/chemokine expression caused by ORMDL3 overexpression. Moreover, in vivo knockdown of ORMDL3 and/or ATF6 enhanced passive cutaneous anaphylaxis (PCA) reactions in mouse ears. These data indicate that ORMDL3 suppresses Ag-mediated mast cell activation via an ATF6 UPR-autophagy dependent pathway and thus, attenuates anaphylactic reaction. This highlights a potential mechanism to intervene in mast cell mediated diseases.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Autofagia , Mastocitos/inmunología , Mastocitos/metabolismo , Proteínas de la Membrana/metabolismo , Transducción de Señal , Respuesta de Proteína Desplegada , Animales , Antígenos/inmunología , Autofagia/inmunología , Degranulación de la Célula/inmunología , Línea Celular , Citocinas/metabolismo , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Inmunomodulación , Proteínas de la Membrana/genética , Ratones , Fosforilación
16.
Clin Exp Pharmacol Physiol ; 37(5-6): 593-7, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20082626

RESUMEN

1. The aim of the present study was to investigate the mechanisms involved in the clearance of isosteviol using the rat isolated perfused liver. 2. Six livers from male Sprague-Dawley rats were perfused with 15.7 mumol isosteviol in a recirculating system. Perfusate and bile samples were collected for 60 min and the liver was collected at the end of the perfusion. All samples collected were incubated with alpha-glucuronidase. Isosteviol-glucuronide was determined as equivalent isosteviol. Isosteviol concentrations were determined using a previously developed liquid chromatography-tandem mass spectrometry method. The final isosteviol liver/perfusate (L/P), bile/liver (B/L) and isosteviol-glucuronide in bile/liver (B(G)/L(G)) ratios were determined. 3. Isosteviol has a high clearance (21.4 +/- 4.8 mL/min) from the perfusate, with a short half-life (13 +/- 4 min). alpha-Glucuronidase incubation revealed that isosteviol is conjugated in the liver and excreted into the bile. There was no isosteviol-glucuronide detected in perfusate samples. The total recovery of the rat isolated perfused liver system is 74 +/- 14% and glucuronidated isosteviol accounted for 23 +/- 4% of the administered dose. 4. In conclusion, we are the first to characterize the metabolism of isosteviol using rat isolated liver perfusion. Our results strongly suggest that the liver is the main organ of isosteviol elimination and that isosteviol is glucuronidated in the liver before it is excreted into the bile.


Asunto(s)
Diterpenos de Tipo Kaurano/farmacocinética , Hígado/metabolismo , Animales , Bilis/metabolismo , Cromatografía Liquida , Diterpenos de Tipo Kaurano/química , Diterpenos de Tipo Kaurano/metabolismo , Glucósidos/metabolismo , Glucurónidos/metabolismo , Glicósido Hidrolasas/farmacología , Técnicas In Vitro , Masculino , Tasa de Depuración Metabólica , Estructura Molecular , Perfusión , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Distribución Tisular
17.
Viruses ; 12(4)2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32276443

RESUMEN

The human immunodeficiency virus type 1 (HIV) establishes a chronic infection that can be well controlled, but not cured, by combined antiretroviral therapy (cART). Interventions have been explored to accomplish a functional cure, meaning that a patient remains infected but HIV is undetectable in the blood, with the aim of allowing patients to live without cART. Tat, the viral transactivator of transcription protein, plays a critical role in controlling HIV transcription, latency, and viral rebound following the interruption of cART treatment. Therefore, a logical approach for controlling HIV would be to block Tat. Tackling Tat with inhibitors has been a difficult task, but some recent discoveries hold promise. Two anti-HIV proteins, Nullbasic (a mutant of Tat) and HT1 (a fusion of HEXIM1 and Tat functional domains) inhibit viral transcription by interfering with the interaction of Tat and cellular factors. Two small molecules, didehydro-cortistatin A (dCA) and triptolide, inhibit Tat by different mechanisms: dCA through direct binding and triptolide through enhanced proteasomal degradation. Finally, two Tat-based vaccines under development elicit Tat-neutralizing antibodies. These vaccines have increased the levels of CD4+ cells and reduced viral loads in HIV-infected people, suggesting that the new vaccines are therapeutic. This review summarizes recent developments of anti-Tat agents and how they could contribute to a functional cure for HIV.


Asunto(s)
Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Animales , Regulación Viral de la Expresión Génica/efectos de los fármacos , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Interacciones Huésped-Patógeno , Humanos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes de Fusión , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Replicación Viral , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
18.
mBio ; 10(4)2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31455650

RESUMEN

Nullbasic is a mutant form of the HIV-1 transcriptional activator protein (Tat) that strongly inhibits HIV-1 transcription and replication in lymphocytes in vitro To investigate Nullbasic inhibition in vivo, we employed an NSG mouse model where animals were engrafted with primary human CD4+ cells expressing a Nullbasic-ZsGreen1 (NB-ZSG) fusion protein or ZSG. NB-ZSG and ZSG were delivered by using a retroviral vector where CD4+ cells were transduced either prior to (preinfection) or following (postinfection) HIV-1 infection. The transduced cells were analyzed in vitro up to 10 days postinfection (dpi) and in vivo up to 39 dpi. Compared to ZSG, NB-ZSG strongly inhibited HIV-1 replication both in vitro and in vivo using preinfection treatment. In vitro, HIV-1 mRNA levels in cells were reduced by up to 60-fold. In vivo, HIV-1 RNA was undetectable in plasma samples during the course of the experiment, and HIV-1 mRNA levels in resident CD4+ cells in organ tissue were reduced up to 2,800-fold. Postinfection treatment of HIV-1-infected cells with NB-ZSG attenuated HIV-1 infection for up to 14 days. In vitro, a 25-fold reduction of viral mRNA in cells was observed but diminished to a <2-fold reduction by 10 dpi. In vivo, HIV-1 RNA was undetectable in plasma of NB-ZSG mice at 14 dpi but afterwards was not significantly different between NB-ZSG mice and control mice. However, we observed higher levels of CD4+ cells in NB-ZSG mice than in control mice, suggesting that NB-ZSG imparted a survival advantage to HIV-1-infected animals.IMPORTANCE HIV-1 infection is effectively controlled by antiviral therapy that inhibits virus replication and reduces viral loads below detectable levels in patients. However, therapy interruption leads to viral rebound due to latently infected cells, which serve as a source of continued viral infection. Interest in strategies leading to a functional cure for HIV-1 infection by long-term or permanent viral suppression is growing. Here, we show that a mutant form of the HIV-1 Tat protein, referred to as Nullbasic, inhibits HIV-1 transcription in infected CD4+ cells in vivo Analysis shows that stable expression of Nullbasic in CD4+ cells could lead to durable anti-HIV-1 activity. Nullbasic, as a gene therapy candidate, could be a part of a functional-cure strategy to suppress HIV-1 transcription and replication.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , VIH-1/fisiología , Proteínas Mutantes/farmacología , Replicación Viral/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Animales , Linfocitos T CD4-Positivos/virología , Infecciones por VIH/virología , VIH-1/genética , Humanos , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Carga Viral , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
19.
Virology ; 530: 65-74, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30782564

RESUMEN

The eukaryotic translation elongation factor 1A (eEF1A) has two cell-type specific paralogs, eEF1A1 and eEF1A2. Both paralogs undertake a canonical function in delivering aminoacyl-tRNA to the ribosome for translation, but differences in other functions are emerging. eEF1A1 has been reported to be important for the replication of many viruses, but no study has specifically linked the eEF1A2 paralog. We have previously demonstrated that eEF1A1 directly interacts with HIV-1 RT and supports efficient reverse transcription. Here, we showed that RT interacted more strongly with eEF1A1 than with eEF1A2 in immunoprecipitation assay. Biolayer interferometry using eEF1A paralogs showed different association and dissociation rates with RT. Over expressed eEF1A1, but not eEF1A2, was able to restore HIV-1 reverse transcription efficiency in HEK293T cells with endogenous eEF1A knocked-down and HIV-1 reverse transcription efficiency correlated with the level of eEF1A1 mRNA, but not to eEF1A2 mRNA in both HEK293T and primary human skeletal muscle cells.


Asunto(s)
Transcriptasa Inversa del VIH/metabolismo , VIH-1/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Factor 1 de Elongación Peptídica/metabolismo , Transcripción Reversa , Células HEK293 , Humanos , Inmunoprecipitación , Células Musculares , Unión Proteica
20.
J Pharm Biomed Anal ; 48(3): 986-90, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18701231

RESUMEN

A sensitive liquid chromatographic-tandem mass spectrometric (LC-MS/MS) method was developed to investigate isosteviol pharmacokinetics in vivo. Isosteviol was extracted from plasma with hexane and 4% formic acid. A Phenomenex Synergi 2mu Fusion reversed phase analytical HPLC column (50 mm x 2.0 mm) equipped with a Synergi 2micro Fusion guard column was employed for chromatographic separations. The gradient mobile phase consisted of acetonitrile (ACN) and 20mM ammonium acetate at pH 6.5, starting at 20% ACN and ramping to 80% at 7 min, followed by 80% ACN for 1 min, then 20% ACN for 5 min. Negative SRM was used to monitor the m/z 317.1/317.1 and 317.3/317.3 transitions for isosteviol and 395.0/395.0 and 397.0/397.0 transitions for internal standard. The retention time of isosteviol was 9.2 min. The assay was linear over the range of 50-2,000 ng/mL. The accuracy of the method was in the range of 97-105%. Intra- and inter-day precisions were in the range of 1.5-4.6%. Isosteviol (4 mg/kg) was dosed intravenously and orally to Sprague-Dawley rats (n=6). Plasma samples were collected and analysed. Intravenous isosteviol has a distribution half-life of 35.7+/-9.0 min with the initial distribution volume of 68.1+/-9.4 mL. The total clearance, terminal half-life and steady-state volume of distribution were 1.25+/-0.12 mL/min, 150.6+/-50.5 min and 272.6+/-95.9 mL, respectively. The oral bioavailability of isosteviol was found to be 60.4+/-15.5%.


Asunto(s)
Cromatografía Liquida/métodos , Diterpenos de Tipo Kaurano/farmacocinética , Edulcorantes/farmacocinética , Espectrometría de Masas en Tándem/métodos , Acetatos/química , Acetonitrilos/química , Administración Oral , Animales , Área Bajo la Curva , Disponibilidad Biológica , Calibración , Cromatografía Liquida/instrumentación , Diterpenos de Tipo Kaurano/administración & dosificación , Diterpenos de Tipo Kaurano/química , Estabilidad de Medicamentos , Congelación , Semivida , Concentración de Iones de Hidrógeno , Inyecciones Intravenosas , Masculino , Tasa de Depuración Metabólica , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Estándares de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Solventes/química , Edulcorantes/administración & dosificación , Edulcorantes/química , Espectrometría de Masas en Tándem/instrumentación , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA