Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Biosci ; 12(1): 77, 2022 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-35641966

RESUMEN

BACKGROUND: Activating transcription factor-2 (ATF2) is a member of the basic leucine zipper family of DNA-binding proteins, which exhibits both oncogenic and tumor suppression activity in different tumors. However, the molecular mechanism of its dual function in cancer chemotherapy especially in gastric cancer has still not been elucidated. METHODS: The protein expression and location of ATF2 in gastric cancer tissues was detected with immunohistochemistry assay, and the clinical significance was analyzed using TCGA and GEO database. The activation and impact of ATF2 in cisplatin treated cells were evaluated with western blot, incucyte live cell analysis, clone formation and tumor xenografts assays. Interaction between ATF2 and p53 was confirmed with immunoprecipitation and GST-pull down. Potential molecular mechanism of ATF2 in different p53 status cells was analyzed with RNA sequencing and real-time quantitative PCR. RESULTS: ATF2 mainly located in the nucleus of cancer cells, higher ATF2 level was associated with poor five-year survival of gastric patients, especially in those undergone chemotherapy treatment. Cisplatin treatment significantly activated ATF2 in p53 mutant cells. ATF2 could interact with the trans-activation domain of p53 and enhance cisplatin sensitivity in p53 wild type cell lines, while promoted cell survival in mutant p53 cancer cells by affecting ERK1/2 pathway. CONCLUSIONS: This study confirmed the effect of ATF2 on cisplatin sensitivity was associated with the functional status of p53 in gastric cancer cells. Integrated analysis of ATF2 expression and P53 status could be used to evaluate the chemotherapy sensitivity and prognosis of gastric cancer patients.

3.
J Immunother Cancer ; 8(1)2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32217766

RESUMEN

BACKGROUND: Local recurrence and remote metastasis are major challenges to overcome in order to improve the survival of patients with cancer after surgery. Oncolytic viruses are a particularly attractive option for prevention of postsurgical disease as they offer a non-toxic treatment option that can directly target residual tumor deposits and beneficially modulate the systemic immune environment that is suppressed post surgery and allows residual disease escape from control. Here, we report that a novel Vaccinia virus (VV), VVΔTKΔN1L (with deletion of both thymidine kinase (TK) and N1L genes) armed with interleukin 12 (IL-12), can prolong postoperative survival when used as a neoadjuvant treatment in different murine and hamster surgical models of cancer. METHODS: A tumor-targeted replicating VV with deletion of TK gene and N1L gene (VVΔTKΔN1L) was created. This virus was armed rationally with IL-12. The effect of VVΔTKΔN1L and VVΔTKΔN1L-IL12 on modulation of the tumor microenvironment and induction of tumor-specific immunity as well the feasibility and safety as a neoadjuvant agent for preventing recurrence and metastasis after surgery were assessed in several clinically relevant models. RESULTS: VVΔTKΔN1L can significantly prolong postoperative survival when used as a neoadjuvant treatment in three different surgery-induced metastatic models of cancer. Efficacy was critically dependent on elevation of circulating natural killer cells that was achieved by virus-induced cytokine production from cells infected with N1L-deleted, but not N1L-intact VV. This effect was further enhanced by arming VVΔTKΔN1L with IL-12, a potent antitumor cytokine. Five daily treatments with VVΔTKΔN1L-IL12 before surgery dramatically improved postsurgical survival. VVΔTKΔN1L armed with human IL-12 completely prevented tumor recurrence in surgical models of head and neck cancer in Syrian hamsters. CONCLUSIONS: These data provide a proof of concept for translation of the regime into clinical trials. VVΔTKΔN1L-IL12 is a promising agent for use as an adjuvant to surgical treatment of solid tumors.


Asunto(s)
Inmunidad/inmunología , Neoplasias Pulmonares/prevención & control , Recurrencia Local de Neoplasia/prevención & control , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Neoplasias Pancreáticas/prevención & control , Virus Vaccinia/genética , Adyuvantes Inmunológicos/administración & dosificación , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Interleucina-12/administración & dosificación , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/cirugía , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/cirugía , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Células Tumorales Cultivadas , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Clin Cancer Res ; 26(2): 465-476, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31767564

RESUMEN

PURPOSE: Pancreatic cancer remains one of the most lethal cancers, and late detection renders most tumors refractory to conventional therapies. Development of cancer prophylaxis may be the most realistic option for improving mortality associated with this disease. Here, we develop a novel individualized prophylactic and therapeutic vaccination regimen using induced pluripotent stem cells (iPSC), gene editing, and tumor-targeted replicating oncolytic viruses. EXPERIMENTAL DESIGN: We created a Virus-Infected, Reprogrammed Somatic cell-derived Tumor cell (VIReST) regime. iPSCs from healthy cells were induced to pancreatic tumor cells using in situ gene editing via stable provision of KRas G12D and p53 R172H tumor driver mutations. These cells were preinfected with oncolytic Adenovirus (AdV) as prime or Vaccinia virus (VV) as boost, to improve vaccine immunogenicity, prior to delivery of vaccines in a sequential regime to young KPC transgenic mice, genetically programmed to develop pancreatic cancer, to prevent and delay disease development. RESULTS: Tumor cells preinfected with oncolytic AdV as prime or VV as boost were the best regime to induce tumor-specific immunity. iPSC-derived tumor cells were highly related in antigen repertoire to pancreatic cancer cells of KPC transgenic mice, suggesting that an individual's stem cells can provide an antigenically matched whole tumor cell vaccine. The VIReST vaccination primed tumor-specific T-cell responses, resulting in delayed disease emergence and progression and significantly prolonged survival of KPC transgenic mice. Importantly, this regime was well-tolerated and nontoxic. CONCLUSIONS: These results provide both proof of concept and a robust technology platform for the development of personalized prophylactic cancer vaccines to prevent pancreatic malignancies in at-risk individuals.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Células Madre Pluripotentes Inducidas/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Experimentales/prevención & control , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Neoplasias Pancreáticas/prevención & control , Animales , Vacunas contra el Cáncer/inmunología , Chlorocebus aethiops , Progresión de la Enfermedad , Masculino , Ratones , Ratones Transgénicos , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Tasa de Supervivencia , Linfocitos T/inmunología , Resultado del Tratamiento
5.
Front Immunol ; 10: 2329, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31632404

RESUMEN

Infectious diseases still remain one of the biggest challenges for human health. In order to gain a better understanding of the pathogenesis of infectious diseases and develop effective diagnostic tools, therapeutic agents, and preventive vaccines, a suitable animal model which can represent the characteristics of infectious is required. The Syrian hamster immune responses to infectious pathogens are similar to humans and as such, this model is advantageous for studying pathogenesis of infection including post-bacterial, viral and parasitic pathogens, along with assessing the efficacy and interactions of medications and vaccines for those pathogens. This review summarizes the current status of Syrian hamster models and their use for understanding the underlying mechanisms of pathogen infection, in addition to their use as a drug discovery platform and provides a strong rationale for the selection of Syrian hamster as animal models in biomedical research. The challenges of using Syrian hamster as an alternative animal model for the research of infectious diseases are also addressed.


Asunto(s)
Enfermedades Transmisibles/etiología , Animales , Enfermedades Transmisibles/diagnóstico , Enfermedades Transmisibles/terapia , Cricetinae , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Interacciones Huésped-Parásitos , Interacciones Huésped-Patógeno , Investigación
6.
Nat Commun ; 9(1): 203, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29321470

RESUMEN

The originally published version of this Article contained errors in Figure 4. In panel b, the square and diamond labels associated with the uppermost survival curve were incorrectly displayed as 'n' and 'u', respectively. These errors have now been corrected in both the PDF and HTML versions of the Article.

7.
Nat Commun ; 8(1): 1395, 2017 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-29123084

RESUMEN

Interleukin-12 (IL-12) has emerged as one of the most potent agents for anti-tumor immunotherapy. However, potentially lethal toxicity associated with systemic administration of IL-12 precludes its clinical application. Here we redesign the molecule in such a way that its anti-tumor efficacy is not compromised, but toxic effects are eliminated. Deletion of the N-terminal signal peptide of IL-12 can effect such a change by preventing IL-12 secretion from cells. We use a newly designed tumor-targeted oncolytic adenovirus (Ad-TD) to deliver non-secreting (ns) IL-12 to tumor cells and examine the therapeutic and toxic effects in Syrian hamster models of pancreatic cancer (PaCa). Strikingly, intraperitoneal delivery of Ad-TD-nsIL-12 significantly enhanced survival of animals with orthotopic PaCa and cured peritoneally disseminated PaCa with no toxic side effects, in contrast to the treatment with Ad-TD expressing unmodified IL-12. These findings offer renewed hope for development of IL-12-based treatments for cancer.


Asunto(s)
Antineoplásicos/farmacología , Inmunoterapia/métodos , Interleucina-12/inmunología , Interleucina-12/farmacología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Adenoviridae/genética , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/química , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Cricetinae , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Humanos , Interleucina-12/efectos adversos , Interleucina-12/química , Neoplasias Pancreáticas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Vis Exp ; (116)2016 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-27842362

RESUMEN

The CRISPR-associated endonuclease Cas9 can edit particular genomic loci directed by a single guide RNA (gRNA). The CRISPR/Cas9 system has been successfully employed for editing genomes of various organisms. Here we describe a protocol for editing the vaccinia virus (VV) genome in the cytoplasm of VV-infected CV-1 cells using the RNA-guided Cas9. RNA-guided Cas9 induces double-stranded DNA breaks facilitating homologous recombination efficiently and specifically in the targeted site of VV and a transgene can be incorporated into these sites by homologous recombination. By using a site-specific homologous vector with transgene(s), a N1L gene-deleted VV with the red fluorescence protein (RFP) gene incorporated in this region was generated with a successful recombination efficiency 10 times greater than that obtained from the conventional homologous recombination method. This protocol demonstrates successful use of RNA-guided Cas9 system to generate mutant VVs with enhanced efficiency.


Asunto(s)
Sistemas CRISPR-Cas , Roturas del ADN de Doble Cadena , Virus Vaccinia , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , ARN Guía de Kinetoplastida
9.
Mol Ther Methods Clin Dev ; 2: 15035, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26417609

RESUMEN

The current method for creation of vaccinia virus (VACV) vectors involves using a selection and purification marker, however inclusion of a gene without therapeutic value in the resulting vector is not desirable for clinical use. The Cre-LoxP system has been used to make marker-free Poxviruses, but the efficiency was very low. To obtain a marker-free VACV vector, we developed marker gene excision systems to modify the thymidine kinase (TK) region and N1L regions using Cre-Loxp and Flp-FRET systems respectively. CRISPR-Cas9 system significantly resulted in a high efficiency (~90%) in generation of marker gene-positive TK-mutant VACV vector. The marker gene (RFP) could be excised from the recombinant virus using Cre recombinase. To make a marker-free VV vector with double gene deletions targeting the TK and N1L gene, we constructed a donor repair vector targeting the N1L gene, which can carry a therapeutic gene and the marker (RFP) that could be excised from the recombinant virus using Flp recombinase. The marker-free system developed here can be used to efficiently construct VACV vectors armed with any therapeutic genes in the TK region or N1L region without marker genes. Our marker-free system platform has significant potential for development of new marker-free VACV vectors for clinical application.

10.
Oncoimmunology ; 4(9): e1038689, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26405610

RESUMEN

Although the profile of safety of tumor-targeted oncolytic virus (TOV) is encouraging, the antitumor efficacy of TOV alone is disappointing. Interleukin-10 (IL-10) plays an important role in carcinogenesis and anti-virus immunity. Here we report that tumor-targeted oncolytic vaccinia virus (VV) armed with IL10 shows promising potential for treatment of pancreatic cancer (PaCa).

11.
J Virol ; 89(9): 5176-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25741005

RESUMEN

Vaccinia virus (VACV) continues to be used in immunotherapy for the prevention of infectious diseases and treatment of cancer since its use for the eradication of smallpox. However, the current method of editing the VACV genome is not efficient. Here, we demonstrate that the CRISPR-Cas9 system can be used to edit the VACV genome rapidly and efficiently. Additionally, a set of 8,964 computationally designed unique guide RNAs (gRNAs) targeting all VACV genes will be valuable for the study of VACV gene functions.


Asunto(s)
Sistemas CRISPR-Cas , Genoma Viral , Biología Molecular/métodos , Recombinación Genética , Virus Vaccinia/genética , Tecnología Farmacéutica/métodos
12.
Clin Cancer Res ; 21(2): 405-16, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25416195

RESUMEN

PURPOSE: Vaccinia virus has strong potential as a novel therapeutic agent for treatment of pancreatic cancer. We investigated whether arming vaccinia virus with interleukin-10 (IL10) could enhance the antitumor efficacy with the view that IL10 might dampen the host immunity to the virus, increasing viral persistence, thus maximizing the oncolytic effect and antitumor immunity associated with vaccinia virus. EXPERIMENTAL DESIGN: The antitumor efficacy of IL10-armed vaccinia virus (VVLΔTK-IL10) and control VVΔTK was assessed in pancreatic cancer cell lines, mice bearing subcutaneous pancreatic cancer tumors and a pancreatic cancer transgenic mouse model. Viral persistence within the tumors was examined and immune depletion experiments as well as immunophenotyping of splenocytes were carried out to dissect the functional mechanisms associated with the viral efficacy. RESULTS: Compared with unarmed VVLΔTK, VVLΔTK-IL10 had a similar level of cytotoxicity and replication in vitro in murine pancreatic cancer cell lines, but rendered a superior antitumor efficacy in the subcutaneous pancreatic cancer model and a K-ras-p53 mutant-transgenic pancreatic cancer model after systemic delivery, with induction of long-term antitumor immunity. The antitumor efficacy of VVLΔTK-IL10 was dependent on CD4(+) and CD8(+), but not NK cells. Clearance of VVLΔTK-IL10 was reduced at early time points compared with the control virus. Treatment with VVLΔTK-IL10 resulted in a reduction in virus-specific, but not tumor-specific CD8(+) cells compared with VVLΔTK. CONCLUSIONS: These results suggest that VVLΔTK-IL10 has strong potential as an antitumor therapeutic for pancreatic cancer.


Asunto(s)
Interleucina-10/genética , Virus Oncolíticos/genética , Neoplasias Pancreáticas/terapia , Virus Vaccinia/genética , Animales , Línea Celular Tumoral , Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias , Viroterapia Oncolítica , Neoplasias Pancreáticas/inmunología , Replicación Viral
13.
J Virol ; 87(5): 2781-90, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23269798

RESUMEN

Vaccinia virus (VV) is an enveloped DNA virus from the poxvirus family and has played a crucial role in the eradication of smallpox. It continues to be used in immunotherapy for the prevention of infectious diseases and treatment of cancer. However, the mechanisms of poxvirus entry, the host factors that affect viral virulence, and the reasons for its natural tropism for tumor cells are incompletely understood. By studying the effect of hypoxia on VV infection, we found that vascular endothelial growth factor A (VEGF-A) augments oncolytic VV cytotoxicity. VEGF derived from tumor cells acts to increase VV internalization, resulting in increased replication and cytotoxicity in an AKT-dependent manner in both tumor cells and normal respiratory epithelial cells. Overexpression of VEGF also enhances VV infection within tumor tissue in vivo after systemic delivery. These results highlight the importance of VEGF expression in VV infection and have potential implications for the design of new strategies to prevent poxvirus infection and the development of future generations of oncolytic VV in combination with conventional or biological therapies.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/metabolismo , Virus Vaccinia/patogenicidad , Factor A de Crecimiento Endotelial Vascular/metabolismo , Internalización del Virus , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/virología , Línea Celular Tumoral , Células Epiteliales/virología , Genes Reporteros , Humanos , Hipoxia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Interferencia de ARN , ARN Interferente Pequeño , Mucosa Respiratoria/virología , Vaccinia/metabolismo , Vaccinia/virología , Virus Vaccinia/genética , Factor A de Crecimiento Endotelial Vascular/genética , Tropismo Viral , Replicación Viral/genética
14.
Clin Cancer Res ; 18(24): 6679-89, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23091113

RESUMEN

PURPOSE: The efficacy of oncolytic viruses depends on multiple actions including direct tumor lysis, modulation of tumor perfusion, and stimulation of tumor-directed immune responses. In this study, we investigated whether a sequential combination of immunologically distinct viruses might enhance antitumor efficacy through the induction of tumor-specific immunity and circumvention or mitigation of antiviral immune responses. EXPERIMENTAL DESIGN: The Syrian hamster as an immune-competent model that supports replication of both adenovirus and vaccinia virus was evaluated in vitro and in vivo. The antitumor efficacy of either virus alone or sequential combination of the two viruses was examined in pancreatic and kidney cancer models. The functional mechanism of the regimen developed here was investigated by histopathology, immunohistochemistry staining, CTL assay, and T-cell depletion. RESULTS: The Syrian hamster is a suitable model for assessment of oncolytic adenovirus and vaccinia virus. Three low doses of adenovirus followed by three low doses of vaccinia virus resulted in a superior antitumor efficacy to the reverse combination, or six doses of either virus alone, against pancreatic and kidney tumors in Syrian hamsters. A total of 62.5% of animals bearing either tumor type treated with the sequential combination became tumor-free, accompanied by the induction of effective tumor-specific immunity. This enhanced efficacy was ablated by CD3+ T-cell depletion but was not associated with humoral immunity against the viruses. CONCLUSION: These findings show that sequential treatment of tumors with oncolytic adenovirus and vaccinia virus is a promising approach for cancer therapy and that T-cell responses play a critical role.


Asunto(s)
Neoplasias Renales/terapia , Viroterapia Oncolítica , Neoplasias Pancreáticas/terapia , Adenoviridae/genética , Adenoviridae/inmunología , Adenoviridae/fisiología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Apoptosis , Células Cultivadas , Cricetinae , Femenino , Humanos , Inmunocompetencia , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Mesocricetus , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Virus Oncolíticos/fisiología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/virología , Carga Tumoral , Virus Vaccinia/genética , Virus Vaccinia/inmunología , Virus Vaccinia/fisiología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Virus Res ; 136(1-2): 192-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18541325

RESUMEN

Peste-des-petits ruminants virus (PPRV) causes acute febrile illness in both farmed and wild small ruminants, with associated mortality rates of 50-80%. PPRV is a member of the Morbillivirus genus within the Paramyxovirus family and although there are many full length genome sequences available for members of this family, their availability for PPRV in particular is limited. We have determined the full length sequences representing two virulent strains of PPRV, the Côte d'Ivoire 1989 (CI/89) and Nigeria 1976 (Ng76/1) strains. We present an alignment of the promoter regions of these viruses with other available PPRV promoter sequences and have identified domains in PPRV proteins believed to be critical for paramyxovirus promoter attenuation. We have also analysed the proteins of these viruses, comparing them to other available PPRV protein sequences and identified motifs that were previously recognised as being required for the function of other paramyxovirus proteins.


Asunto(s)
Genoma Viral , Virus de la Peste de los Pequeños Rumiantes/genética , ARN Viral/genética , Animales , Secuencia de Bases , Côte d'Ivoire , Datos de Secuencia Molecular , Nigeria , Virus de la Peste de los Pequeños Rumiantes/aislamiento & purificación , Regiones Promotoras Genéticas , Rumiantes , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Proteínas Virales/genética
16.
Virus Res ; 126(1-2): 250-5, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17350130

RESUMEN

Peste-des-petits-ruminants virus (PPRV) (family Paramyxoviridae, genus Morbillivirus) causes an acute febrile illness in sheep and goats resulting in significant morbidity and mortality in infected herds. The paramyxoviruses all have negative sense, non-segmented RNA genomes and their host range and pathogenic determinants have been extensively studied using reverse genetics. This technology also enables a more rational approach to be taken with respect to vaccine design. In order to initiate this type of work for PPRV we constructed a PPRV minigenome and studied its expression in transfected cells. As for other morbilliviruses, the minimum requirements for minigenome rescue were shown to be the cis-acting elements of the genome (GP) and antigenome (AGP) promoters as well as the three trans-acting helper proteins N (nucleocapsid), P (phosphoprotein) and L (large polymerase). Homologous PPRV helper proteins were compared to their heterologous analogues from the closely related rinderpest virus (RPV) and heterologous minigenome rescue was found to be a much less efficient process. By engineering two GP/AGP chimeric minigenomes we also identified differences between the two viruses in the specific interactions between the promoters and the transcriptase/replicase complexes. The PPRV minigenome was also shown not to strictly comply with the "rule of six"in vitro.


Asunto(s)
Virus de la Peste de los Pequeños Rumiantes/genética , Regiones Promotoras Genéticas , Animales , Quimera/genética , Técnicas Genéticas , Genoma Viral , Cabras , Virus Helper/genética , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/patogenicidad , Plásmidos/genética , Virus de la Peste Bovina/genética , Ovinos , Proteínas Virales/genética
17.
J Gen Virol ; 87(Pt 4): 927-936, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16528042

RESUMEN

The internal ribosome entry site (IRES) elements from porcine enterovirus 8 and simian virus 2, two members of a proposed new genus within the family Picornaviridae, were characterized. These IRES elements, in common with the porcine teschovirus 1 IRES, were found to be related functionally and structurally to the IRES element from Hepatitis C virus, a member of the family Flaviviridae. Partial secondary structure predictions were derived and functional assays demonstrated that these IRES elements continued to be active when eIF4G was cleaved and when the activity of eIF4A was blocked.


Asunto(s)
Enterovirus Porcinos/genética , Hepacivirus/genética , Picornaviridae/genética , Picornaviridae/patogenicidad , Ribosomas/metabolismo , Regiones no Traducidas 5'/química , Regiones no Traducidas 5'/genética , Animales , Secuencia de Bases , Codón Iniciador , ADN Complementario/química , ADN Complementario/genética , Enterovirus Porcinos/patogenicidad , Hepacivirus/patogenicidad , Humanos , Datos de Secuencia Molecular , Picornaviridae/clasificación , ARN Viral/química , ARN Viral/genética , Porcinos/virología
18.
Nat Chem Biol ; 2(4): 213-20, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16532013

RESUMEN

RNA helicases are molecular motors that are involved in virtually all aspects of RNA metabolism. Eukaryotic initiation factor (eIF) 4A is the prototypical member of the DEAD-box family of RNA helicases. It is thought to use energy from ATP hydrolysis to unwind mRNA structure and, in conjunction with other translation factors, it prepares mRNA templates for ribosome recruitment during translation initiation. In screening marine extracts for new eukaryotic translation initiation inhibitors, we identified the natural product hippuristanol. We show here that this compound is a selective and potent inhibitor of eIF4A RNA-binding activity that can be used to distinguish between eIF4A-dependent and -independent modes of translation initiation in vitro and in vivo. We also show that poliovirus replication is delayed when infected cells are exposed to hippuristanol. Our study demonstrates the feasibility of selectively targeting members of the DEAD-box helicase family with small-molecule inhibitors.


Asunto(s)
Factor 4A Eucariótico de Iniciación/química , ARN Helicasas/química , Ribosomas/química , Adenosina Trifosfato/química , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/farmacología , Relación Dosis-Respuesta a Droga , Escherichia coli/metabolismo , Factor 4A Eucariótico de Iniciación/metabolismo , Humanos , Hidrólisis , Espectroscopía de Resonancia Magnética , Modelos Genéticos , Plásmidos/metabolismo , Poliovirus/genética , Poliovirus/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Biosíntesis de Proteínas , ARN/química , ARN Mensajero/metabolismo , Proteínas Recombinantes/química , Relación Estructura-Actividad , Transfección
19.
J Virol ; 80(3): 1271-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16415004

RESUMEN

The internal ribosome entry site (IRES) of porcine teschovirus 1 (PTV-1), a member of the Picornaviridae family, is quite distinct from other well-characterized picornavirus IRES elements, but it displays functional similarities to the IRES from hepatitis C virus (HCV), a member of the Flaviviridae family. In particular, a dominant negative mutant form of eIF4A does not inhibit the activity of the PTV-1 IRES. Furthermore, there is a high level (ca. 50%) of identity between the PTV-1 and HCV IRES sequences. A secondary-structure model of the whole PTV-1 IRES has been derived which includes a pseudoknot. Validation of specific features within the model has been achieved by mutagenesis and functional assays. The differences and similarities between the PTV-1 and HCV IRES elements should assist in defining the critical features of this type of IRES.


Asunto(s)
Enterovirus Porcinos/genética , Hepacivirus/genética , ARN Viral/química , ARN Viral/genética , Animales , Secuencia de Bases , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación de Ácido Nucleico , ARN Mensajero/química , ARN Mensajero/genética , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Porcinos
20.
J Virol ; 78(9): 4487-97, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15078929

RESUMEN

Initiation of protein synthesis on picornavirus RNA requires an internal ribosome entry site (IRES). Typically, picornavirus IRES elements contain about 450 nucleotides (nt) and use most of the cellular translation initiation factors. However, it is now shown that just 280 nt of the porcine teschovirus type 1 Talfan (PTV-1) 5' untranslated region direct the efficient internal initiation of translation in vitro and within cells. In toeprinting assays, assembly of 48S preinitiation complexes from purified components on the PTV-1 IRES was achieved with just 40S ribosomal subunits plus eIF2 and Met-tRNA(i)(Met). Indeed, a binary complex between 40S subunits and the PTV-1 IRES is formed. Thus, the PTV-1 IRES has properties that are entirely different from other picornavirus IRES elements but highly reminiscent of the hepatitis C virus (HCV) IRES. Comparison between the PTV-1 IRES and HCV IRES elements revealed islands of high sequence identity that occur in regions critical for the interactions of the HCV IRES with the 40S ribosomal subunit and eIF3. Thus, there is significant functional and structural similarity between the IRES elements from the picornavirus PTV-1 and HCV, a flavivirus.


Asunto(s)
Hepacivirus/patogenicidad , Picornaviridae/patogenicidad , Ribosomas/metabolismo , Regiones no Traducidas 5' , Animales , Secuencia de Bases , Codón Iniciador , Hepacivirus/genética , Datos de Secuencia Molecular , Picornaviridae/genética , Biosíntesis de Proteínas , ARN Viral/química , ARN Viral/metabolismo , Proteínas Ribosómicas/química , Proteínas Ribosómicas/metabolismo , Porcinos/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...