Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Viruses ; 16(7)2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-39066190

RESUMEN

Negative-strand RNA viruses form cytoplasmic inclusion bodies (IBs) representing virus replication foci through phase separation or biomolecular condensation of viral and cellular proteins, as a hallmark of their infection. Alternatively, mammalian cells form stalled mRNA containing antiviral stress granules (SGs), as a consequence of phosphorylation of eukaryotic initiation factor 2α (eIF2α) through condensation of several RNA-binding proteins including TIA-1. Whether and how Chandipura virus (CHPV), an emerging human pathogen causing influenza-like illness, coma and death, forms IBs and evades antiviral SGs remain unknown. By confocal imaging on CHPV-infected Vero-E6 cells, we found that CHPV infection does not induce formation of distinct canonical SGs. Instead, CHPV proteins condense and co-localize together with SG proteins to form heterogeneous IBs, which ensued independent of the activation of eIF2α and eIF2α kinase, protein kinase R (PKR). Interestingly, siRNA-mediated depletion of PKR or TIA-1 significantly decreased viral transcription and virion production. Moreover, CHPV infection also caused condensation and recruitment of PKR to IBs. Compared to SGs, IBs exhibited significant rapidity in disassembly dynamics. Altogether, our study demonstrating that CHPV replication co-optimizes with SG proteins and revealing an unprecedented proviral role of TIA-1/PKR may have implications in understanding the mechanisms regulating CHPV-IB formation and designing antiviral therapeutics. Importance: CHPV is an emerging tropical pathogen reported to cause acute influenza-like illness and encephalitis in children with a very high mortality rate of ~70%. Lack of vaccines and an effective therapy against CHPV makes it a potent pathogen for causing an epidemic in tropical parts of globe. Given these forewarnings, it is of paramount importance that CHPV biology must be understood comprehensively. Targeting of host factors offers several advantages over targeting the viral components due to the generally higher mutation rate in the viral genome. In this study, we aimed at understanding the role of SGs forming cellular RNA-binding proteins in CHPV replication. Our study helps understand participation of cellular factors in CHPV replication and could help develop effective therapeutics against the virus.


Asunto(s)
Cuerpos de Inclusión Viral , Antígeno Intracelular 1 de las Células T , Replicación Viral , eIF-2 Quinasa , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Animales , Antígeno Intracelular 1 de las Células T/metabolismo , Antígeno Intracelular 1 de las Células T/genética , Chlorocebus aethiops , Células Vero , Cuerpos de Inclusión Viral/metabolismo , Humanos , Gránulos de Estrés/metabolismo , Cuerpos de Inclusión/metabolismo , Interacciones Huésped-Patógeno , Gránulos Citoplasmáticos/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/genética , Separación de Fases
2.
Mol Cell Biochem ; 2023 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-37285039

RESUMEN

CUL4A is an ubiquitin ligase deregulated in numerous pathologies including cancer and even hijacked by viruses for facilitating their survival and propagation. However, its role in Human papilloma virus (HPV)-mediated cervical carcinogenesis remains elusive. The UALCAN and GEPIA datasets were analyzed to ascertain the transcript levels of CUL4A in cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) patients. Subsequently, various biochemical assays were employed to explore the functional contribution of CUL4A in cervical carcinogenesis and to shed some light on its involvement in Cisplatin resistance in cervical cancer. Our UALCAN and GEPIA datasets analyses reveal elevated CUL4A transcript levels in cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) patients that correlate with adverse clinicopathological parameters such as tumor stage and lymph node metastasis. Kaplan-Meier plot and GEPIA assessment depict poor prognosis of CESC patients having high CUL4A expression. Varied biochemical assays illustrate that CUL4A inhibition severely curtails hallmark malignant properties such as cellular proliferation, migration, and invasion of cervical cancer cells. We also show that CUL4A knockdown in HeLa cells causes increased susceptibility and better apoptotic induction toward Cisplatin, a mainstay drug used in cervical cancer treatment. More interestingly, we find reversion of Cisplatin-resistant phenotype of HeLa cells and an augmented cytotoxicity towards the platinum compound upon CUL4A downregulation. Taken together, our study underscores CUL4A as a cervical cancer oncogene and illustrates its potential as a prognosis indicator. Our investigation provides a novel avenue in improving current anti-cervical cancer therapy and overcoming the bottle-neck of Cisplatin resistance.

3.
J Am Chem Soc ; 145(1): 579-584, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36524964

RESUMEN

An amphiphilic segmented polyurethane (F-PU-S), with pendant sulfate groups and a flexible hydrocarbon backbone, exhibits intrachain H-bonding-reinforced folding and hierarchical assembly, producing an anionic polymersome with efficient display of sulfate groups at the surface. It shows an excellent antiviral activity against Sendai virus (SV) by inhibiting its entry to the cells. Mechanistic investigation suggests fusion of the SV and the polymersome to produce larger particles in which neither the folded structure of the polymer nor the fusogenic property of the SV exists anymore. In sharp contrast, a structurally similar polymer R-PU-S, in which the chain folding pathway is blocked by replacing the flexible C6 chain with a rigid cyclohexane chain in the backbone, cannot form a similar polymersome structure and hence does not exhibit any antiviral activity. On the other hand, the third polymer (F-PU-C), which is similar to F-PU-S except for the pendant anionic groups (carboxylate instead of sulfate), also fails to exhibit any antiviral activity against SV, confirming the essential role of the chain folding as well as the pendant sulfate groups for the fusion-induced antiviral activity of F-PU-S, which provides an important structural guideline for developing new antiviral polymers.


Asunto(s)
Polímeros , Polímeros/farmacología , Estructura Secundaria de Proteína
4.
Sci Rep ; 11(1): 13253, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34168211

RESUMEN

Chandipura virus (CHPV, a member of the Rhabdoviridae family) is an emerging pathogen that causes rapidly progressing influenza-like illness and acute encephalitis often leading to coma and death of the human host. Given several CHPV outbreaks in Indian sub-continent, recurring sporadic cases, neurological manifestation, and high mortality rate of this infection, CHPV is gaining global attention. The 'dark proteome' includes the whole proteome with special emphasis on intrinsically disordered proteins (IDP) and IDP regions (IDPR), which are proteins or protein regions that lack unique (or ordered) three-dimensional structures within the cellular milieu. These proteins/regions, however, play a number of vital roles in various biological processes, such as cell cycle regulation, control of signaling pathways, etc. and, therefore, are implicated in many human diseases. IDPs and IPPRs are also abundantly found in many viral proteins enabling their multifunctional roles in the viral life cycles and their capability to highjack various host systems. The unknown abundance of IDP and IDPR in CHPV, therefore, prompted us to analyze the dark proteome of this virus. Our analysis revealed a varying degree of disorder in all five CHPV proteins, with the maximum level of intrinsic disorder propensity being found in Phosphoprotein (P). We have also shown the flexibility of P protein using extensive molecular dynamics simulations up to 500 ns (ns). Furthermore, our analysis also showed the abundant presence of the disorder-based binding regions (also known as molecular recognition features, MoRFs) in CHPV proteins. The identification of IDPs/IDPRs in CHPV proteins suggests that their disordered regions may function as potential interacting domains and may also serve as novel targets for disorder-based drug designs.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/metabolismo , Infecciones por Rhabdoviridae/metabolismo , Vesiculovirus/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Genoma Viral/genética , Humanos , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Fosfoproteínas/metabolismo , Proteoma , Infecciones por Rhabdoviridae/virología , Alineación de Secuencia , Vesiculovirus/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
5.
Front Microbiol ; 12: 618559, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33959102

RESUMEN

Understanding the structure-function of inclusion bodies (IBs) in the last two decades has led to the development of several mild solubilization buffers for the improved recovery of bioactive proteins. The recently developed freeze-thaw-based inclusion body protein solubilization method has received a great deal of attention due to its simplicity and cost-effectiveness. The present report investigates the reproducibility, efficiency, and plausible mechanism of the freeze-thaw-based IB solubilization. The percentage recovery of functionally active protein species of human growth hormone (hGH) and L-asparaginase from their IBs in Escherichia coli and the quality attributes associated with the freeze-thaw-based solubilization method were analyzed in detail. The overall yield of the purified hGH and L-asparaginase protein was found to be around 14 and 25%, respectively. Both purified proteins had functionally active species lower than that observed with commercial proteins. Biophysical and biochemical analyses revealed that the formation of soluble aggregates was a major limitation in the case of tough IB protein like hGH. On the other hand, the destabilization of soft IB protein like L-asparaginase led to the poor recovery of functionally active protein species. Our study provides insight into the advantages, disadvantages, and molecular-structural information associated with the freeze-thaw-based solubilization method.

6.
Viruses ; 12(9)2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32825063

RESUMEN

COVID-19 novel coronavirus (CoV) disease caused by severe acquired respiratory syndrome (SARS)-CoV-2 manifests severe lethal respiratory illness in humans and has recently developed into a worldwide pandemic. The lack of effective treatment strategy and vaccines against the SARS-CoV-2 poses a threat to human health. An extremely high infection rate and multi-organ secondary infection within a short period of time makes this virus more deadly and challenging for therapeutic interventions. Despite high sequence similarity and utilization of common host-cell receptor, human angiotensin-converting enzyme-2 (ACE2) for virus entry, SARS-CoV-2 is much more infectious than SARS-CoV. Structure-based sequence comparison of the N-terminal domain (NTD) of the spike protein of Middle East respiratory syndrome (MERS)-CoV, SARS-CoV, and SARS-CoV-2 illustrate three divergent loop regions in SARS-CoV-2, which is reminiscent of MERS-CoV sialoside binding pockets. Comparative binding analysis with host sialosides revealed conformational flexibility of SARS-CoV-2 divergent loop regions to accommodate diverse glycan-rich sialosides. These key differences with SARS-CoV and similarity with MERS-CoV suggest an evolutionary adaptation of SARS-CoV-2 spike glycoprotein reciprocal interaction with host surface sialosides to infect host cells with wide tissue tropism.


Asunto(s)
Betacoronavirus/química , Coronavirus del Síndrome Respiratorio de Oriente Medio/química , Ácidos Siálicos/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Amino Azúcares/metabolismo , Betacoronavirus/fisiología , Sitios de Unión , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Ácido N-Acetilneuramínico/metabolismo , Unión Proteica , Dominios Proteicos , Receptores de Coronavirus , Receptores Virales/química , Receptores Virales/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , SARS-CoV-2 , Antígeno Sialil Lewis X/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Tropismo Viral , Internalización del Virus
7.
FEBS Lett ; 593(15): 1927-1943, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31183850

RESUMEN

Connivance of cellular factors during virus-host cell membrane fusion is poorly understood. We have recently shown that cellular villin plays an important role during membrane fusion of reconstituted Sendai virosomes with hepatocytes. Here, we employed villin-null Chinese Hamster Ovary (CHO) cells, where villin expression led to an increased fusion with virosomes, which was further enhanced due to tyrosine phosphorylation in the presence of c-src. However, the villin RRI mutant, lacking actin-severing function, failed to augment membrane fusion. Furthermore, quantitative mass spectrometry and detailed analysis revealed Tyr499 to be the key phosphorylation site of villin responsible for the enhancement of virosome-CHO cell fusion. Overall, our results demonstrate a critical role for villin and its cell-type dependent phosphorylation in regulating membrane fusion.


Asunto(s)
Membrana Celular/virología , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Virus Sendai/fisiología , Citoesqueleto de Actina/metabolismo , Animales , Células CHO , Membrana Celular/fisiología , Cricetulus , Interacciones Huésped-Patógeno , Fusión de Membrana , Proteínas de Microfilamentos/metabolismo , Mutación , Fosforilación , Tirosina/química , Familia-src Quinasas/metabolismo
8.
Appl Microbiol Biotechnol ; 102(14): 6221-6234, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29855689

RESUMEN

Protein expression in the milk of transgenic farmed animals offers a cost-effective system for producing therapeutics. However, transgenesis in farmed animals is not only cumbersome but also involves risk of potential hazard by germline gene integration, due to interruptions caused by the transgene in the native genome. Avoiding germline gene integration, we have delivered buffalo ß-casein promoter-driven transgene construct entrapped in virosomes directly in the milk gland through intraductal perfusion delivery. Virosomes were generated from purified Sendai viral membrane, containing hemagglutinin-neuraminidase (HN) and fusion factor (F) proteins on surface (HNF-Virosomes) which initiate membrane fusion, devoid of any viral nucleic acids. Intraductal delivery of HNF-Virosomes predominantly transfected luminal epithelial cells lining the milk duct and buffalo ß-casein promoter of the construct ensured mammary luminal epithelial cell specific expression of the transgene. Mammary epithelial cells expressed EGFP at lactation when egfp was used as a transgene. Similarly, human interferon-γ (hIFN-γ) was expressed in the mammary gland as well as in the milk when hIFN-γ was used as a transgene. This combinatorial approach of using Sendai viral membrane-derived virosomes for entrapment and delivery of the transgene and using buffalo ß-casein promoter for mammary gland specific gene expression provided a better option for generating therapeutic proteins in milk, bypassing germline gene integration avoiding risks associated with animal bioreactor generated through germline gene integration.


Asunto(s)
Terapia Biológica/métodos , Búfalos/genética , Expresión Génica/genética , Lactancia/genética , Glándulas Mamarias Animales/metabolismo , Leche/química , Transgenes/genética , Animales , Caseínas/genética , Femenino , Humanos , Regiones Promotoras Genéticas/genética , Virus Sendai/genética
9.
Mol Biol Cell ; 28(26): 3801-3814, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29074568

RESUMEN

Reconstituted Sendai viral envelopes (virosomes) are well recognized for their promising potential in membrane fusion-mediated delivery of bioactive molecules to liver cells. Despite the known function of viral envelope glycoproteins in catalyzing fusion with cellular membrane, the role of host cell proteins remains elusive. Here, we used two-dimensional differential in-gel electrophoresis to analyze hepatic cells in early response to virosome-induced membrane fusion. Quantitative mass spectrometry together with biochemical analysis revealed that villin, an actin-modifying protein, is differentially up-regulated and phosphorylated at threonine 206-an early molecular event during membrane fusion. We found that villin influences actin dynamics and that this influence, in turn, promotes membrane mixing through active participation of Sendai viral envelope glycoproteins. Modulation of villin in host cells also resulted in a discernible effect on the entry and egress of progeny Sendai virus. Taken together, these results suggest a novel mechanism of regulated viral entry in animal cells mediated by host factor villin.


Asunto(s)
Hepatocitos/metabolismo , Fusión de Membrana/fisiología , Proteínas de Microfilamentos/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/fisiología , Animales , Membrana Celular/metabolismo , Células HeLa , Células Hep G2 , Hepatocitos/fisiología , Humanos , Proteínas de Microfilamentos/fisiología , Virus Sendai/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Virosomas/metabolismo
10.
Methods Mol Biol ; 1543: 245-257, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28349432

RESUMEN

Double-stranded RNA-mediated transcriptional gene silencing (TGS) has shown promising results over posttranscriptional gene silencing (PTGS) due to its long term and heritable nature. Various research groups have shed light on different mechanisms by which TGS operate. Some of these include histone modification, DNA methylation, or restriction of RNA polymerase binding onto the target gene's promoter. This serves as an added advantage since permanent c-Myc inactivation is critical for suppressing hepatocellular carcinoma (HCC). Inability to target cancer cells specifically, without affecting the normal cells, has been one of the biggest drawbacks of an effective cancer therapy. Therefore, we aimed to overcome this barrier by first generating tumor-specific transcriptional units expressing TGS inducing shRNAs against c-Myc's P2 promoter only in neoplastic liver cells. Secondly, we coupled this TGS inducing system with Sendai fusion virosomes for liver-specific delivery to minimize nonspecific side effects in vitro.


Asunto(s)
Carcinoma Hepatocelular/genética , Silenciador del Gen , Técnicas de Transferencia de Gen , Genes myc , Neoplasias Hepáticas/genética , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Línea Celular Tumoral , Células Cultivadas , Islas de CpG , Metilación de ADN , Humanos , ARN Interferente Pequeño/administración & dosificación , Transfección/métodos , Virosomas
12.
J Transl Med ; 13: 254, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26242403

RESUMEN

BACKGROUND: Placental like alkaline phosphatase (PLAP), an oncofetal antigen, is highly expressed in germ cell, cervical, ovarian and several other tumour types but minimally in normal tissues [corrected]. The expression of a PLAP promoter based transcriptional unit following antigen mediated cell specific delivery is a possible approach for tumour targeting. METHODS: PLAP promoter alone or in combination with NFκB DNA response elements was used for expressing shRNA targeting the long control region (LCR) of human papillomavirus (HPV)-16 oncogenes E6 and E7 via transcriptional gene silencing in PLAP expressing cervical cancer cell lines, SiHa and CaSki. This was packaged in a Sendai virus envelope incorporating a single chain variable fragment antibody (scFv) for antibody mediated targeting. Specificity and efficacy of the shRNA was assessed by studying the heterochromatization, down regulation of the HPV-16 E6/E7 genes and subsequent effects on their targets and cell growth properties. RESULTS: Reduction of HPV-16 E6 and E7 expression by TGS led to the activation of the previously suppressed target genes of p53 (PUMA and NOXA) and Rb (cyclins A2 and E). Cell death was seen only in PLAP expressing HPV-16 infected SiHa and CaSki cells but not in the HPV-18 integrated HeLa and non-PLAP CHO cells. There was reduction in the enhancer associated transcripts of the long control region (LCR) of HPV-16 E6/E7 genes. Also, an increase in the enrichment of dimethylated histone three lysine nine (H3K9Me2) and trimethylated histone three lysine twenty-seven (H3K27Me3) was observed by ChIP assay, which decreased upon trichostatin A treatment, indicating a possible mechanism for the heterochromatization of the target LCR region. CONCLUSION: A combination of novel PLAP promoter and antibody based specificities has the potential for being developed as a possible therapeutic strategy for PLAP positive neoplasia.


Asunto(s)
Fosfatasa Alcalina/genética , Silenciador del Gen , Técnicas de Transferencia de Gen , Isoenzimas/genética , Neoplasias/metabolismo , Regiones Promotoras Genéticas , Anticuerpos de Cadena Única/metabolismo , Virosomas/metabolismo , Apoptosis , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN/genética , Factor de Transcripción E2F1/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación Neoplásica de la Expresión Génica , Papillomavirus Humano 16/metabolismo , Humanos , Cinética , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
13.
Sci Rep ; 5: 10395, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-25993465

RESUMEN

Enveloped viruses enter host cells through membrane fusion and the cells in turn alter their shape to accommodate components of the virus. However, the role of nonmuscle myosin II of the actomyosin complex of host cells in membrane fusion is yet to be understood. Herein, we show that both (-) blebbistatin, a specific inhibitor of nonmuscle myosin II (NMII) and small interfering RNA markedly augment fusion of Sendai virus (SeV), with chinese hamster ovary cells and human hepatocarcinoma cells. Inhibition of RLC phosphorylation using inhibitors against ROCK, but not PKC and MRCK, or overexpression of phospho-dead mutant of RLC enhances membrane fusion. SeV infection increases cellular stiffness and myosin light chain phosphorylation at two hour post infection. Taken together, the present investigation strongly indicates that Rho-ROCK-NMII contractility signaling pathway may provide a physical barrier to host cells against viral fusion.


Asunto(s)
Miosina Tipo IIA no Muscular/metabolismo , Virus Sendai/fisiología , Secuencia de Aminoácidos , Animales , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Microscopía de Fuerza Atómica , Microscopía Fluorescente , Mutagénesis , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Miosina Tipo IIA no Muscular/antagonistas & inhibidores , Miosina Tipo IIA no Muscular/genética , Miosina Tipo IIB no Muscular/metabolismo , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Alineación de Secuencia , Internalización del Virus/efectos de los fármacos , Liberación del Virus/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
14.
BMC Cancer ; 14: 582, 2014 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-25108398

RESUMEN

BACKGROUND: A specific targeting modality for hepatocellular carcinoma (HCC) could ideally encompass a liver cell specific delivery system of a transcriptional unit that is active only in neoplastic cells. Sendai virosomes, derived from Sendai viral envelopes, home to hepatocytes based on the liver specific expression of asialoglycoprotein receptors (ASGPRs) which are recognized by the Sendai virosomal fusion (F) proteins. As reported earlier by us and other groups, transcriptional gene silencing (TGS) does not require continuous presence of the effector siRNA/shRNA molecule and is heritable, involving epigenetic modifications, leading to long term transcriptional repression. This could be advantageous over conventional gene therapy approaches, since continuous c-Myc inactivation is required to suppress hepatocarcinoma cells. METHODS: Exploiting such virosomal delivery, the alpha-fetoprotein (AFP) promoter, in combination with various tumour specific enhancers, was used to drive the expression of shRNA directed against ME1a1 binding site of the proto-oncogene c-Myc P2 promoter, in order to induce TGS in neoplastic liver cells. RESULTS: The dual specificity achieved by the Sendai virosomal delivery system and the promoter/enhancer guided expression ensured that the shRNA inducing TGS was active only in liver cells that had undergone malignant transformation. Our results indicate that such a bimodal therapeutic system induced specific activation of apoptosis in hepatocarcinoma cells due to heterochromatization and increased DNA methylation of the CpG islands around the target loci. CONCLUSIONS: The Sendai virosomal delivery system, combined with AFP promoter/enhancer expression machinery, could serve as a generalized mechanism for the expression of genes deleterious to transformed hepatocarcinoma cells. In this system, the epigenetic suppression of c-Myc could have an added advantage for inducing cell death in the targeted cells.


Asunto(s)
Carcinoma Hepatocelular/genética , Hepatocitos/metabolismo , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogénicas c-myc/genética , alfa-Fetoproteínas/genética , Animales , Células CHO , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Islas de CpG , Cricetulus , Metilación de ADN , Silenciador del Gen , Terapia Genética , Células Hep G2 , Hepatocitos/virología , Humanos , Neoplasias Hepáticas/patología , Especificidad de Órganos , Regiones Promotoras Genéticas , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Interferente Pequeño/genética , Virus Sendai/genética , Virosomas
15.
Mol Ther ; 21(1): 57-67, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22910295

RESUMEN

Recently, we have demonstrated that the protease domain of NS3 alone can bind specifically to hepatitis C virus (HCV) internal ribosome entry site (IRES) near the initiator AUG, dislodges human La protein and inhibits translation in favor of viral RNA replication. Here, by using a computational approach, the contact points of the protease on the HCV IRES were putatively mapped. A 30-mer NS3 peptide was designed from the predicted RNA-binding region that retained RNA-binding ability and also inhibited IRES-mediated translation. This peptide was truncated to 15 mer and this also demonstrated ability to inhibit HCV RNA-directed translation as well as replication. More importantly, its activity was tested in an in vivo mouse model by encapsulating the peptide in Sendai virus virosomes followed by intravenous delivery. The study demonstrates for the first time that the HCV NS3-IRES RNA interaction can be selectively inhibited using a small peptide and reports a strategy to deliver the peptide into the liver.


Asunto(s)
Péptidos/farmacología , Ribosomas/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Sitios de Unión , Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Humanos , Ratones , Datos de Secuencia Molecular , Biosíntesis de Proteínas , ARN Viral/genética , ARN Viral/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Ribosomas/metabolismo , Homología de Secuencia de Aminoácido , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
16.
RNA Biol ; 9(8): 1110-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22858675

RESUMEN

Translation initiation of hepatitis C Virus (HCV) RNA is the initial obligatory step of the viral life cycle, mediated through the Internal Ribosome Entry Site (IRES) present in the 5'-untranslated region (UTR). Initiation on the HCV IRES is mediated by multiple structure-specific interactions between IRES RNA and host 40S ribosomal subunit. In the present study we demonstrate that the SLIIIef domain, in isolation from other structural elements of HCV IRES, retain the ability to interact with 40S ribosome subunit. A small RNA SLRef, mimicking the SLIIIef domain was found to interact specifically with human La protein and the ribosomal protein S5 and selectively inhibit HCV RNA translation. More importantly, SLRef RNA showed significant suppression of replication in HCV monocistronic replicon and decrease of negative strand synthesis in HCV cell culture system. Finally, using Sendai virus based virosome, the targeted delivery of SLRef RNA into mice liver succeeded in selectively inhibiting HCV IRES mediated translation in vivo.


Asunto(s)
Hepacivirus/genética , Iniciación de la Cadena Peptídica Traduccional/efectos de los fármacos , ARN Pequeño no Traducido/farmacología , ARN Viral/metabolismo , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , Animales , Secuencia de Bases , Femenino , Hepacivirus/metabolismo , Hígado , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , ARN Pequeño no Traducido/química , Replicación Viral/efectos de los fármacos
17.
J Virol ; 84(9): 4366-82, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20164223

RESUMEN

Viruses of the Paramyxoviridae family bind to their host cells by using hemagglutinin-neuraminidase (HN), which enhances fusion protein (F)-mediated membrane fusion. Although respiratory syncytial virus and parainfluenza virus 5 of this family are suggested to trigger host cell signaling during infection, the virus-induced intracellular signals dictating virus-cell fusion await elucidation. Using an F- or HN-F-containing reconstituted envelope of Sendai virus, another paramyxovirus, we revealed the role and regulation of AKT1 and Raf/MEK/ERK cascades during viral fusion with liver cells. Our observation that extracellular signal-regulated kinase (ERK) activation promotes viral fusion via ezrin-mediated cytoskeletal rearrangements, whereas AKT1 attenuates fusion by promoting phosphorylation of F protein, indicates a counteractive regulation of viral fusion by reciprocal activation of AKT1 and mitogen-activated protein kinase (MAPK) cascades, establishing a novel conceptual framework for a therapeutic strategy.


Asunto(s)
Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Virus Sendai/fisiología , Transducción de Señal , Internalización del Virus , Línea Celular , Proteína HN/genética , Proteína HN/metabolismo , Hepatocitos/virología , Humanos , Virus Sendai/genética , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo
18.
Hepatology ; 50(3): 815-24, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19585550

RESUMEN

UNLABELLED: Asialoglycoprotein receptor (ASGPR)-mediated endocytosis has been used to target genes to hepatocytes in vivo. However, the level and duration of transgene expression have been low because of lysosomal translocation and degradation of the DNA and lack of its integration into the host genome. In this study we packaged the DNA of interest in proteoliposomes containing the fusogenic galactose-terminated F-glycoprotein of the Sendai virus (FPL) for targeted delivery to hepatocytes. After the FPL binds to ASGPR on the hepatocyte surface, fusogenic activity of the F-protein delivers the DNA into the cytosol, bypassing the endosomal pathway. For transgene integration we designed plasmids containing one transcription unit expressing the Sleeping Beauty transposase (SB) and another expressing human uridinediphosphoglucuronate glucuronosyltransferase-1A1 (pSB-hUGT1A1). The latter was flanked by inverted/direct repeats that are substrates of SB. In cell culture, FPL-mediated delivery of the E. coli beta-galactosidase gene (LacZ) resulted in transduction of ASGPR-positive cells (rat hepatocytes or Hepa1 cell line), but not of ASGPR-negative 293 cells. Intravenous injection of the FPL-entrapped pSB-hUGT1A1 (4-8 microg/day, 1-4 doses) into UGT1A1-deficient hyperbilirubinemic Gunn rats (model of Crigler-Najjar syndrome type 1) resulted in hUGT1A1 expression in 5%-10% of hepatocytes, but not in other cell types. Serum bilirubin levels declined by 30% +/- 4% in 2 weeks and remained at that level throughout the 7-month study duration. With histidine containing FPL, serum bilirubin was reduced by 40% +/- 5%, and bilirubin glucuronides were excreted into bile. No antibodies were detectable in the recipient rats against the F-protein or human UGT1A1. CONCLUSION: FPL is an efficient hepatocyte-targeted gene delivery platform in vivo that warrants further exploration toward clinical application.


Asunto(s)
Receptor de Asialoglicoproteína/administración & dosificación , Ictericia/terapia , Proteolípidos/administración & dosificación , Transposasas/administración & dosificación , Animales , Síndrome de Crigler-Najjar/terapia , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Glucuronosiltransferasa/administración & dosificación , Hepatocitos/efectos de los fármacos , Humanos , Hiperbilirrubinemia/terapia , Ratas , Ratas Gunn , Proteínas Virales de Fusión/administración & dosificación
19.
J Gen Virol ; 90(Pt 8): 1812-1819, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19297606

RESUMEN

Internal ribosome entry site (IRES)-mediated translation of input viral RNA is the initial required step for the replication of the positive-stranded genome of hepatitis C virus (HCV). We have shown previously the importance of the GCAC sequence near the initiator AUG within the stem and loop IV (SLIV) region in mediating ribosome assembly on HCV RNA. Here, we demonstrate selective inhibition of HCV-IRES-mediated translation using short hairpin (sh)RNA targeting the same site within the HCV IRES. sh-SLIV showed significant inhibition of viral RNA replication in a human hepatocellular carcinoma (Huh7) cell line harbouring a HCV monocistronic replicon. More importantly, co-transfection of infectious HCV-H77s RNA and sh-SLIV in Huh7.5 cells successfully demonstrated a significant decrease in viral RNA in HCV cell culture. Additionally, we report, for the first time, the targeted delivery of sh-SLIV RNA into mice liver using Sendai virosomes and demonstrate selective inhibition of HCV-IRES-mediated translation. Results provide the proof of concept that Sendai virosomes could be used for the efficient delivery of shRNAs into liver tissue to block HCV replication.


Asunto(s)
Antivirales/administración & dosificación , Hepatitis C/tratamiento farmacológico , ARN Interferente Pequeño/administración & dosificación , Animales , Línea Celular Tumoral , Hepacivirus/efectos de los fármacos , Humanos , Hígado/virología , Luciferasas/metabolismo , Masculino , Ratones , ARN Viral/genética , ARN Viral/metabolismo , Virus Sendai/genética , Ensayo de Placa Viral , Virosomas , Replicación Viral/efectos de los fármacos
20.
Hum Immunol ; 70(4): 222-9, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19272325

RESUMEN

We investigated the association of human leukocyte antigen (HLA) II (DRB1 and DQB1) alleles with susceptibility to human papillomavirus (HPV)-associated cervical precancer and cancer cases in a hospital-based case-control study in a northern Indian population. A total of 202 subjects, including 100 patients comprising 31 cervical precancer (cervical intraepithelial neoplasia [CIN] 2/3) and 69 invasive cervical cancer cases, and 102 healthy controls participated in the study. Both patients and controls were screened for HPV infection using a polymerase chain reaction (PCR-based approach. Low-resolution PCR-sequence specific priming (PCR-SSP) was used to genotype HLA II (DRB1 and DQB1). Our results demonstrate that the DRB1*15 allele/DRB1*15-DQB1*06 haplotype may have a predisposition for HPV infection (p(c) < 0.05) or cervical cancer/precancer (p(c) < 0.05) development, whereas the DRB1*04 allele/DRB1*04-DQB1*03 haplotype might exhibit susceptibility to cervical precancerous lesions (p(c) < 0.05). The DRB1*13 allele/DRB1*13-DQB1*06 haplotype was strongly protective against risk to HPV infection (p(c) < 0.002) as well as cervical cancer (p(c) 0.01). Therefore, we have demonstrated that HLA DR-DQ polymorphisms are involved in genetic susceptibility to cervical cancer or HPV infection in a northern Indian population.


Asunto(s)
Antígenos HLA-DQ/genética , Antígenos HLA-DR/genética , Infecciones por Papillomavirus/genética , Displasia del Cuello del Útero/genética , Neoplasias del Cuello Uterino/genética , Adulto , Anciano , Alelos , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Cadenas beta de HLA-DQ , Cadenas HLA-DRB1 , Haplotipos , Humanos , India , Desequilibrio de Ligamiento , Persona de Mediana Edad , Infecciones por Papillomavirus/complicaciones , Reacción en Cadena de la Polimerasa , Polimorfismo Genético , Polimorfismo Conformacional Retorcido-Simple , Neoplasias del Cuello Uterino/complicaciones , Adulto Joven , Displasia del Cuello del Útero/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA