Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Epidemiol ; 32(2): 105-111, 2022 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-34776499

RESUMEN

BACKGROUND: Tokyo, the capital of Japan, is a densely populated city of >13 million people, so the population is at high risk of epidemic severe acute respiratory coronavirus 2 (SARS-CoV-2) infection. A serologic survey of anti-SARS-CoV-2 IgG would provide valuable data for assessing the city's SARS-CoV-2 infection status. Therefore, this cross-sectional study estimated the anti-SARS-CoV-2 IgG seroprevalence in Tokyo. METHODS: Leftover serum of 23,234 hospital visitors was tested for antibodies against SARS-CoV-2 using an iFlash 3000 chemiluminescence immunoassay analyzer (Shenzhen YHLO Biotech, Shenzhen, China) with an iFlash-SARS-CoV-2 IgG kit (YHLO) and iFlash-SARS-CoV-2 IgG-S1 kit (YHLO). Serum samples with a positive result (≥10 AU/mL) in either of these assays were considered seropositive for anti-SARS-CoV-2 IgG. Participants were randomly selected from patients visiting 14 Tokyo hospitals between September 1, 2020 and March 31, 2021. No participants were diagnosed with coronavirus disease 2019 (COVID-19), and none exhibited COVID-19-related symptoms at the time of blood collection. RESULTS: The overall anti-SARS-CoV-2 IgG seroprevalence among all participants was 1.83% (95% confidence interval [CI], 1.66-2.01%). The seroprevalence in March 2021, the most recent month of this study, was 2.70% (95% CI, 2.16-3.34%). After adjusting for population age, sex, and region, the estimated seroprevalence in Tokyo was 3.40%, indicating that 470,778 individuals had a history of SARS-CoV-2 infection. CONCLUSIONS: The estimated number of individuals in Tokyo with a history of SARS-CoV-2 infection was 3.9-fold higher than the number of confirmed cases. Our study enhances understanding of the SARS-CoV-2 epidemic in Tokyo.


Asunto(s)
COVID-19 , Anticuerpos Antivirales , Estudios Transversales , Hospitales , Humanos , Inmunoglobulina G , SARS-CoV-2 , Estudios Seroepidemiológicos , Tokio/epidemiología
2.
J Biol Chem ; 294(15): 5759-5773, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755480

RESUMEN

Hepatitis C virus (HCV) establishes a chronic infection that can lead to cirrhosis and hepatocellular carcinoma. The HCV life cycle is closely associated with host factors that promote or restrict viral replication, the characterization of which could help to identify potential therapeutic targets. To this end, here we performed a genome-wide microarray analysis and identified ribonucleotide reductase M2 (RRM2) as a cellular factor essential for HCV replication. We found that RRM2 is up-regulated in response to HCV infection in quiescent hepatocytes from humanized chimeric mouse livers. To elucidate the molecular basis of RRM2 expression in HCV-infected cells, we used HCV-infected hepatocytes from chimeric mice and hepatoma cells infected with the HCV strain JFH1. Both models exhibited increased RRM2 mRNA and protein expression levels. Moreover, siRNA-mediated silencing of RRM2 suppressed HCV replication and infection. Of note, RRM2 and RNA polymerase nonstructural protein 5B (NS5B) partially co-localized in cells and co-immunoprecipitated, suggesting that they might interact. RRM2 knockdown reduced NS5B expression, which depended on the protein degradation pathway, as NS5B RNA levels did not decrease and NS5B protein stability correlated with RRM2 protein levels. We also found that RRM2 silencing decreased levels of hPLIC1 (human homolog 1 of protein linking integrin-associated protein and cytoskeleton), a ubiquitin-like protein that interacts with NS5B and promotes its degradation. This finding suggests that there is a dynamic interplay between RRM2 and the NS5B-hPLIC1 complex that has an important function in HCV replication. Together, these results identify a role of host RRM2 in viral RNA replication.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Hepacivirus/fisiología , Hepatitis C Crónica/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ribonucleósido Difosfato Reductasa/biosíntesis , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Regulación Enzimológica de la Expresión Génica , Hepatitis C Crónica/genética , Hepatitis C Crónica/patología , Humanos , Hígado/metabolismo , Hígado/patología , Hígado/virología , Ratones , Ratones SCID , Ratones Transgénicos , Complejo de la Endopetidasa Proteasomal/genética , Estabilidad Proteica , Proteolisis , Ribonucleósido Difosfato Reductasa/genética , Ubiquitinación/genética , Proteínas no Estructurales Virales/genética
3.
J Hepatol ; 64(3): 547-55, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26505121

RESUMEN

BACKGROUND & AIMS: Antiviral agents including entecavir (ETV) suppress the replication of the hepatitis B virus (HBV) genome in human hepatocytes, but they do not reduce the abundance of viral proteins. The present study focused on effectively reducing viral protein levels. METHODS: We designed siRNAs (HBV-siRNA) that target consensus sequences in HBV genomes. To prevent the emergence of escaped mutant virus, we mixed three HBV-siRNAs (HBV-siRNAmix); the mixture was encapsulated in a novel pH-sensitive multifunctional envelope-type nanodevice (MEND), a hepatocyte-specific drug delivery system. Coagulation factor 7 siRNA was used to assess delivery and knockdown efficiencies of MEND/siRNA treatments in mice. The potency of MEND/HBV-siRNAmix was evaluated in primary human hepatocytes and in chimeric mice with humanized liver persistently infected with HBV. RESULTS: Effective knockdown of targets, efficient delivery of siRNA, and liver-specific delivery were each observed with MEND. MEND/HBV-siRNA caused efficient reduction of HBsAg and HBeAg in vitro and in vivo. However, ETV treatment did not efficiently reduce HBsAg or HBeAg when compared with a single MEND/HBV-siRNAmix treatment. Furthermore, the suppressive effects of a single dose of MEND/HBV-siRNAmix persisted for 14days in vitro and in vivo. CONCLUSION: We demonstrated that MEND/HBV-siRNA controlled HBV more efficiently than did ETV. Furthermore, the effect of a single dose of MEND/HBV-siRNA persisted for a long time. These results indicated that MEND/HBV-siRNA may be a promising novel HBV treatment that is more effective than reverse transcriptase inhibitors.


Asunto(s)
Técnicas de Transferencia de Gen , Hepatitis B Crónica/terapia , ARN Interferente Pequeño/administración & dosificación , Animales , ADN Viral/análisis , Antígenos de Superficie de la Hepatitis B/análisis , Antígenos e de la Hepatitis B/análisis , Virus de la Hepatitis B/genética , Humanos , Concentración de Iones de Hidrógeno , Liposomas , Ratones
4.
J Virol ; 89(8): 4092-101, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25673715

RESUMEN

UNLABELLED: The course of hepatitis C virus (HCV) infection and disease progression involves alterations in lipid metabolism, leading to symptoms such as hypocholesterolemia and steatosis. Steatosis can be induced by multiple mechanisms, including increases in lipid biosynthesis and uptake, impaired lipoprotein secretion, and/or attenuation of lipid ß-oxidation. However, little is known about the effects of HCV on lipid ß-oxidation. A previous proteomics study revealed that HCV interacted with both the α- and ß-subunits of the mitochondrial trifunctional protein (MTP), an enzyme complex which catalyzes the last 3 steps of mitochondrial lipid ß-oxidation for cellular energy production. Here we show that in HCV-infected Huh7.5 cells, lipid ß-oxidation was significantly attenuated. Consistently with this, MTP protein and mRNA levels were suppressed by HCV infection. A loss-of-function study showed that MTP depletion rendered cells less responsive to alpha interferon (IFN-α) treatment by impairing IFN-stimulated gene expression. These aspects of host-virus interaction explain how HCV alters host energy homeostasis and how it may also contribute to the establishment of persistent infection in the liver. IMPORTANCE: HCV infection triggers metabolic alterations, which lead to significant disease outcomes, such as fatty liver (steatosis). This study revealed that HCV impairs mitochondrial lipid ß-oxidation, which results in low lipid combustion. On the other hand, the HCV-induced defects in metabolic status played an important role in the control of the type I interferon system. Under the conditions of impaired lipid ß-oxidation, host cells were less responsive to the ability of exogenously added IFN-α to suppress HCV replication. This suggests that interference with lipid ß-oxidation may assist the virus in the establishment of a long-term, persistent infection. Further understanding of this aspect of virus-host interaction may lead to improvements in the current standard therapy.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hepacivirus/metabolismo , Hepatitis C/metabolismo , Proteína Trifuncional Mitocondrial/metabolismo , Western Blotting , Línea Celular Tumoral , Vectores Genéticos/genética , Interacciones Huésped-Patógeno , Humanos , Interferón-alfa/metabolismo , Metabolismo de los Lípidos/fisiología , Luciferasas , Oxidación-Reducción , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Am J Pathol ; 184(1): 171-83, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24200852

RESUMEN

Patients infected with highly pathogenic avian influenza A H5N1 viruses (H5N1 HPAIV) show diffuse alveolar damage. However, the temporal progression of tissue damage and repair after viral infection remains poorly defined. Therefore, we assessed the sequential histopathological characteristics of mouse lung after intranasal infection with H5N1 HPAIV or H1N1 2009 pandemic influenza virus (H1N1 pdm). We determined the amount and localization of virus in the lung through IHC staining and in situ hybridization. IHC used antibodies raised against the virus protein and antibodies specific for macrophages, type II pneumocytes, or proliferating cell nuclear antigen. In situ hybridization used RNA probes against both viral RNA and mRNA encoding the nucleoprotein and the hemagglutinin protein. H5N1 HPAIV infection and replication were observed in multiple lung cell types and might result in rapid progression of lung injury. Both type II pneumocytes and macrophages proliferated after H5N1 HPAIV infection. However, the abundant macrophages failed to block the viral attack, and proliferation of type II pneumocytes failed to restore the damaged alveoli. In contrast, mice infected with H1N1 pdm exhibited modest proliferation of type II pneumocytes and macrophages and slight alveolar damage. These results suggest that the virulence of H5N1 HPAIV results from the wide range of cell tropism of the virus, excessive virus replication, and rapid development of diffuse alveolar damage.


Asunto(s)
Células Epiteliales Alveolares/virología , Subtipo H5N1 del Virus de la Influenza A/fisiología , Macrófagos/virología , Infecciones por Orthomyxoviridae/patología , Neumonía Viral/patología , Neumonía Viral/virología , Células Epiteliales Alveolares/patología , Animales , Modelos Animales de Enfermedad , Inmunohistoquímica , Hibridación in Situ , Subtipo H1N1 del Virus de la Influenza A , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología , Replicación Viral/fisiología
6.
Gastroenterology ; 145(4): 865-73, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23791700

RESUMEN

BACKGROUND & AIMS: Host cell lipid rafts form a scaffold required for replication of hepatitis C virus (HCV). Serine palmitoyltransferases (SPTs) produce sphingolipids, which are essential components of the lipid rafts that associate with HCV nonstructural proteins. Prevention of the de novo synthesis of sphingolipids by an SPT inhibitor disrupts the HCV replication complex and thereby inhibits HCV replication. We investigated the ability of the SPT inhibitor NA808 to prevent HCV replication in cells and mice. METHODS: We tested the ability of NA808 to inhibit SPT's enzymatic activity in FLR3-1 replicon cells. We used a replicon system to select for HCV variants that became resistant to NA808 at concentrations 4- to 6-fold the 50% inhibitory concentration, after 14 rounds of cell passage. We assessed the ability of NA808 or telaprevir to inhibit replication of HCV genotypes 1a, 1b, 2a, 3a, and 4a in mice with humanized livers (transplanted with human hepatocytes). NA808 was injected intravenously, with or without pegylated interferon alfa-2a and HCV polymerase and/or protease inhibitors. RESULTS: NA808 prevented HCV replication via noncompetitive inhibition of SPT; no resistance mutations developed. NA808 prevented replication of all HCV genotypes tested in mice with humanized livers. Intravenous NA808 significantly reduced viral load in the mice and had synergistic effects with pegylated interferon alfa-2a and HCV polymerase and protease inhibitors. CONCLUSIONS: The SPT inhibitor NA808 prevents replication of HCV genotypes 1a, 1b, 2a, 3a, and 4a in cultured hepatocytes and in mice with humanized livers. It might be developed for treatment of HCV infection or used in combination with pegylated interferon alfa-2a or HCV polymerase or protease inhibitors.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Hepatocitos/virología , Serina C-Palmitoiltransferasa/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Animales , Hepacivirus/clasificación , Hepacivirus/genética , Humanos , Ratones , ARN Viral/análisis
7.
Sci Rep ; 12(1): 4150, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35264719

RESUMEN

Models of animals that are susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can usefully evaluate the efficacy of vaccines and therapeutics. In this study, we demonstrate that infection with the SARS-CoV-2 B.1.351 variant (TY8-612 strain) induces bodyweight loss and inflammatory cytokine/chemokine production in wild-type laboratory mice (BALB/c and C57BL/6 J mice). Furthermore, compared to their counterparts, BALB/c mice had a higher viral load in their lungs and worse symptoms. Importantly, infecting aged BALB/c mice (older than 6 months) with the TY8-612 strain elicited a massive and sustained production of multiple pro-inflammatory cytokines/chemokines and led to universal mortality. These results indicated that the SARS-CoV-2 B.1.351 variant-infected mice exhibited symptoms ranging from mild to fatal depending on their strain and age. Our data provide insights into the pathogenesis of SARS-CoV-2 and may be useful in developing prophylactics and therapeutics.


Asunto(s)
COVID-19/patología , SARS-CoV-2/fisiología , Envejecimiento , Animales , COVID-19/mortalidad , COVID-19/virología , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Análisis de Componente Principal , ARN Viral/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Índice de Severidad de la Enfermedad , Carga Viral
8.
Front Microbiol ; 13: 967019, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36466631

RESUMEN

As long as the coronavirus disease-2019 (COVID-19) pandemic continues, new variants of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) with altered antigenicity will emerge. The development of vaccines that elicit robust, broad, and durable protection against SARS-CoV-2 variants is urgently required. We have developed a vaccine consisting of the attenuated vaccinia virus Dairen-I (DIs) strain platform carrying the SARS-CoV-2 S gene (rDIs-S). rDIs-S induced neutralizing antibody and T-lymphocyte responses in cynomolgus macaques and human angiotensin-converting enzyme 2 (hACE2) transgenic mice, and the mouse model showed broad protection against SARS-CoV-2 isolates ranging from the early-pandemic strain (WK-521) to the recent Omicron BA.1 variant (TY38-873). Using a tandem mass tag (TMT)-based quantitative proteomic analysis of lung homogenates from hACE2 transgenic mice, we found that, among mice subjected to challenge infection with WK-521, vaccination with rDIs-S prevented protein expression related to the severe pathogenic effects of SARS-CoV-2 infection (tissue destruction, inflammation, coagulation, fibrosis, and angiogenesis) and restored protein expression related to immune responses (antigen presentation and cellular response to stress). Furthermore, long-term studies in mice showed that vaccination with rDIs-S maintains S protein-specific antibody titers for at least 6 months after a first vaccination. Thus, rDIs-S appears to provide broad and durable protective immunity against SARS-CoV-2, including current variants such as Omicron BA.1 and possibly future variants.

9.
Biochem Biophys Res Commun ; 410(3): 404-9, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21669189

RESUMEN

Nonstructural protein 5A (NS5A) of hepatitis C virus (HCV) plays multiple and diverse roles in the viral lifecycle, and is currently recognized as a novel target for anti-viral therapy. To establish an HCV cell culture system with NS5A of various strains, recombinant viruses were generated by replacing NS5A of strain JFH-1 with those of strains of genotypes 1 (H77; 1a and Con1; 1b) and 2 (J6CF; 2a and MA; 2b). All these recombinant viruses were capable of replication and infectious virus production. The replacement of JFH-1 NS5A with those of genotype 1 strains resulted in similar or slightly reduced virus production, whereas replacement with those of genotype 2 strains enhanced virus production as compared with JFH-1 wild-type. A single cycle virus production assay with a CD81-negative cell line revealed that the efficient virus production elicited by replacement with genotype 2 strains depended on enhanced viral assembly, and that substitutions in the C-terminus of NS5A were responsible for this phenotype. Pulse-chase assays revealed that these substitutions in the C-terminus of NS5A were possibly associated with accelerated cleavage kinetics at the NS5A-NS5B site. Using this cell culture system with NS5A-substituted recombinant viruses, the anti-viral effects of an NS5A inhibitor were then examined. A 300- to 1000-fold difference in susceptibility to the inhibitor was found between strains of genotypes 1 and 2. This system will facilitate not only a better understanding of strain-specific roles of NS5A in the HCV lifecycle, but also enable the evaluation of genotype and strain dependency of NS5A inhibitors.


Asunto(s)
Hepacivirus/genética , Hepacivirus/fisiología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/fisiología , Ensamble de Virus/fisiología , Sustitución de Aminoácidos , Antivirales/farmacología , Carbamatos , Línea Celular Tumoral , Farmacorresistencia Viral/genética , Humanos , Imidazoles/farmacología , Pirrolidinas , Valina/análogos & derivados , Proteínas no Estructurales Virales/antagonistas & inhibidores , Ensamble de Virus/genética
10.
mSphere ; 6(3)2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33980684

RESUMEN

Antibody detection is crucial for monitoring host immune responses to specific pathogen antigens (Ags) and evaluating vaccine efficacies. The luciferase immunoprecipitation system (LIPS) was developed for sensitive detection of Ag-specific antibodies in sera from various species. In this study, we describe NanoLIPS, an improved LIPS assay based on NanoLuciferase (NLuc), and employ the assay for monitoring antibody responses following influenza virus infection or vaccination. We generated recombinant influenza virus hemagglutinin (HA) proteins tagged with N-terminal (N-NLuc-HA) or C-terminal (C-NLuc-HA) NLuc reporters. NLuc-HA yielded an at least 20-fold higher signal-to-noise ratio than did a LIPS assay employing a recombinant HA-Gaussia princeps luciferase (GLuc) fusion protein. NanoLIPS-based detection of anti-HA antibodies yielded highly reproducible results with a broad dynamic range. The levels of antibodies against C-NLuc-HA generated by mice vaccinated with recombinant vaccinia virus DIs strain expressing an influenza virus HA protein (rDIs-HA) was significantly correlated with the protective effect elicited by the rDIs-HA vaccine. C-NLuc-HA underwent glycosylation with native conformations and assembly to form a trimeric structure and was detected by monoclonal antibodies that detect conformational epitopes present on the globular head or stalk domain of HA. Therefore, NanoLIPS is applicable for evaluating vaccine efficacy. We also showed that C-NLuc-HA is applicable for detection of HA-specific antibodies in sera from various experimental species, including mouse, cynomolgus macaque, and tree shrew. Thus, NanoLIPS-based detection of HA offers a simple and high-sensitivity method that detects native conformational epitopes and can be used in various experimental animal models.IMPORTANCE Influenza virus HA-specific antibodies can be detected via the hemagglutination inhibition (HI) assay, the neutralization (NT) assay, and the enzyme-linked immunosorbent assay (ELISA). However, these assays have some drawbacks, including narrow dynamic range and the requirement for large amounts of sera. As an alternative to an ELISA-based method, luciferase immunoprecipitation system (LIPS) was developed. We focused on NanoLuciferase (NLuc), which has a small size, higher intensity, and longer stability. In this study, we developed a technically feasible and highly sensitive method for detecting influenza virus-specific antibodies using a NLuc-tagged recombinant HA protein produced in mammalian cells. HA with a C-terminal NLuc extension (C-NLuc-HA) was glycosylated and formed trimeric complexes when expressed in mammalian cells. Furthermore, C-NLuc-HA was recognized not only by monoclonal antibodies that bind to the globular head domain but also by those that bind to the stalk domain. We also demonstrated that the data obtained by this assay correlate with the protection of an experimental vaccine in animal models.


Asunto(s)
Epítopos/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Inmunoprecipitación/métodos , Inmunoprecipitación/normas , Luciferasas/química , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales , Epítopos/química , Femenino , Pruebas de Inhibición de Hemaglutinación , Inmunoprecipitación/instrumentación , Vacunas contra la Influenza/inmunología , Luciferasas/metabolismo , Macaca fascicularis , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/diagnóstico , Infecciones por Orthomyxoviridae/inmunología , Sensibilidad y Especificidad , Tupaiidae
11.
Nat Commun ; 12(1): 2654, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976181

RESUMEN

Most anti-influenza drugs currently used, such as oseltamivir and zanamivir, inhibit the enzymatic activity of neuraminidase. However, neuraminidase inhibitor-resistant viruses have already been identified from various influenza virus isolates. Here, we report the development of a class of macrocyclic peptides that bind the influenza viral envelope protein hemagglutinin, named iHA. Of 28 iHAs examined, iHA-24 and iHA-100 have inhibitory effects on the in vitro replication of a wide range of Group 1 influenza viruses. In particular, iHA-100 bifunctionally inhibits hemagglutinin-mediated adsorption and membrane fusion through binding to the stalk domain of hemagglutinin. Moreover, iHA-100 shows powerful efficacy in inhibiting the growth of highly pathogenic influenza viruses and preventing severe pneumonia at later stages of infection in mouse and non-human primate cynomolgus macaque models. This study shows the potential for developing cyclic peptides that can be produced more efficiently than antibodies and have multiple functions as next-generation, mid-sized biomolecules.


Asunto(s)
Antivirales/farmacología , Modelos Animales de Enfermedad , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Péptidos/farmacología , Neumonía/prevención & control , Animales , Antivirales/química , Perros , Femenino , Células HEK293 , Humanos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Macaca fascicularis , Células de Riñón Canino Madin Darby , Ratones Endogámicos BALB C , Estructura Molecular , Péptidos/química , Replicación Viral/efectos de los fármacos
13.
PLoS Pathog ; 3(9): 1335-47, 2007 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-17907805

RESUMEN

Hepatitis C virus (HCV) is a positive-strand RNA virus that frequently causes persistent infections and is uniquely associated with the development of hepatocellular carcinoma. While the mechanism(s) by which the virus promotes cancer are poorly defined, previous studies indicate that the HCV RNA-dependent RNA polymerase, nonstructural protein 5B (NS5B), forms a complex with the retinoblastoma tumor suppressor protein (pRb), targeting it for degradation, activating E2F-responsive promoters, and stimulating cellular proliferation. Here, we describe the mechanism underlying pRb regulation by HCV and its relevance to HCV infection. We show that the abundance of pRb is strongly downregulated, and its normal nuclear localization altered to include a major cytoplasmic component, following infection of cultured hepatoma cells with either genotype 1a or 2a HCV. We further demonstrate that this is due to NS5B-dependent ubiquitination of pRb and its subsequent degradation via the proteasome. The NS5B-dependent ubiquitination of pRb requires the ubiquitin ligase activity of E6-associated protein (E6AP), as pRb abundance was restored by siRNA knockdown of E6AP or overexpression of a dominant-negative E6AP mutant in cells containing HCV RNA replicons. E6AP also forms a complex with pRb in an NS5B-dependent manner. These findings suggest a novel mechanism for the regulation of pRb in which the HCV NS5B protein traps pRb in the cytoplasm, and subsequently recruits E6AP to this complex in a process that leads to the ubiquitination of pRb. The disruption of pRb/E2F regulatory pathways in cells infected with HCV is likely to promote hepatocellular proliferation and chromosomal instability, factors important for the development of liver cancer.


Asunto(s)
Hepacivirus/metabolismo , Proteína de Retinoblastoma/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Carcinoma Hepatocelular/etiología , Regulación hacia Abajo , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Interferencia de ARN , ARN Polimerasa Dependiente del ARN/metabolismo , Células Tumorales Cultivadas , Ubiquitina/metabolismo , Proteínas no Estructurales Virales/metabolismo
14.
J Control Release ; 266: 216-225, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-28986168

RESUMEN

Lipid nanoparticles (LNPs) are the leading technology for delivering short interfering RNA (siRNA) in vivo. While numerous attempts to improve the efficiency of siRNA delivery have been reported, only a few studies of the mechanism of LNP-mediated toxicity and attempts to develop safe LNPs in vivo have been reported, in spite of the significance of such systems, in the light of treatment and clinical applications. We herein report on the elucidation of the mechanism of hepatotoxicity following the intravenous injection of a high dose of hepatotropic LNPs. The LNPs accumulated in liver sinusoidal endothelial cells (LSECs), resulting in their activation and the induction of several cytokines related to neutrophils, followed by neutrophilic inflammation. To circumvent this toxic cascade, the LNPs were modified with a hepatocyte-specific ligand, N-acetyl-d-galactosamine (GalNAc), which resulted in a substantial improvement of hepatocyte-specificity and in a dramatic reduction in toxicity. Moreover, modification of the GalNAc-LNPs with polyethyleneglycol abrogated the LNP-associated toxicity without any detectable loss of gene silencing activity in hepatocytes. Finally, we observed that a single injection of the LNPs resulted in a significant reduction of hepatitis B virus (HBV) genomic DNA and their antigens without any sign of toxicity in chimeric mice with humanized livers that had been persistently infected with HBV. These lines of the fact suggest that the newly designed siRNA-loaded LNPs promise to be a useful technology for the treatment of liver diseases.


Asunto(s)
Virus de la Hepatitis B/efectos de los fármacos , Hepatocitos/metabolismo , ARN Interferente Pequeño/administración & dosificación , Acetilgalactosamina/administración & dosificación , Animales , Citocinas/sangre , ADN Viral/sangre , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Factor VII/genética , Femenino , Hepatitis B/sangre , Hepatitis B/tratamiento farmacológico , Hepatitis B/virología , Virus de la Hepatitis B/genética , Lípidos/administración & dosificación , Lípidos/toxicidad , Ratones Endogámicos ICR , Nanopartículas/administración & dosificación , Nanopartículas/toxicidad , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Polietilenglicoles/administración & dosificación
16.
Antiviral Res ; 108: 79-87, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24893207

RESUMEN

Hepatitis C virus (HCV) is a causative agent of chronic hepatitis. Although the standard therapy for HCV-infected patients consists of pegylated interferon plus ribavirin, this treatment is associated with serious side effects and high costs, and fails in some patients infected with specific HCV genotypes. To address this problem, we are developing small-molecule inhibitors of cyclin-dependent kinases (CDKs) as novel anti-HCV drug candidates. Previous data showed that HCV replication is inhibited by retinoblastoma protein, which is itself inactivated by CDK-mediated phosphorylation. Here, we report that CDK inhibitors suppress HCV replication in vitro and in vivo, and that CDK4 is required for efficient HCV replication. These findings shed light on the development of novel anti-HCV drugs that target host factors.


Asunto(s)
Antivirales/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Replicación Viral/efectos de los fármacos , Línea Celular , Descubrimiento de Drogas/métodos , Humanos , Concentración 50 Inhibidora
17.
PLoS One ; 8(11): e76753, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24223117

RESUMEN

Immunochromatography (IC) is an antigen-detection assay that plays an important role in the rapid diagnosis of influenza virus because the protocol is short time and easy to use. Despite the usability of IC, the sensitivity is approximately 10(3) pfu per reaction. In addition, antigen-antibody interaction-based method cannot be used for the detection of influenza viruses with major antigenic change. In this study, we established the use of fluorescent immunochromatography (FLIC) to detect a broad spectrum of H5 subtype influenza A viruses. This method has improved sensitivity 10-100 fold higher than traditional IC because of the use of fluorescent conjugated beads. Our Type-E FLIC kit detected all of the H5 subtype influenza viruses that were examined, as well as recombinant hemagglutinin (HA) proteins (rHAs) belonging to the Eurasian H5 subtype viruses and the Type-N diagnosed North American H5 subtype influenza A viruses. Thus, this kit has the improved potential to detect H5 subtype influenza viruses of different clades with both Type-E and Type-N FLIC kits. Compared with PCR-based diagnosis, FLIC has a strong advantage in usability, because the sample preparation required for FLIC is only mix-and-drop without any additional steps such as RNA extraction. Our results can provide new strategies against the spread and transmission of HPAI H5N1 viruses in birds and mammals including humans.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Gripe Humana/diagnóstico , Animales , Anticuerpos Inmovilizados/química , Anticuerpos Antivirales/química , Especificidad de Anticuerpos , Cromatografía de Afinidad , Perros , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunoensayo , Subtipo H5N1 del Virus de la Influenza A/inmunología , Gripe Humana/virología , Límite de Detección , Células de Riñón Canino Madin Darby , Juego de Reactivos para Diagnóstico , Espectrometría de Fluorescencia
18.
Proc Natl Acad Sci U S A ; 102(50): 18159-64, 2005 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-16332962

RESUMEN

The retinoblastoma tumor-suppressor protein (Rb) plays a critical role in controlling cellular proliferation and apoptosis by regulating E2F transcription factors. Rb is a key target of oncoproteins expressed by DNA tumor viruses, but RNA viruses are not known to regulate Rb function. Here, we show that Rb abundance is negatively regulated in cells containing replicating genomic RNA from hepatitis C virus, a human virus strongly associated with hepatocellular carcinoma. The viral RNA-dependent RNA polymerase NS5B forms a complex with Rb, targeting it for degradation and resulting in reduction of Rb abundance, activation of E2F-responsive promoters, and cell proliferation. NS5B contains a conserved Leu-x-Cys/Asn-x-Asp motif that is homologous to Rb-binding domains in the oncoproteins of DNA viruses. This domain overlaps the polymerase active site, and mutations within it abrogate Rb binding and reverse the effects of NS5B on E2F promoter activation and cell proliferation. These findings suggest a unique link between an oncogenic RNA virus implicated in the development of liver cancer and a critically important tumor-suppressor protein.


Asunto(s)
Regulación hacia Abajo/genética , Hepacivirus/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteínas no Estructurales Virales/metabolismo , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Línea Celular , Factores de Transcripción E2F/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Luciferasas , Datos de Secuencia Molecular , Mutación/genética , Regiones Promotoras Genéticas/genética , Proteínas no Estructurales Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA