Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Immunother Cancer ; 8(1)2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32217766

RESUMO

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.


Assuntos
Imunidade/imunologia , Neoplasias Pulmonares/prevenção & controle , Recidiva Local de Neoplasia/prevenção & controle , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Neoplasias Pancreáticas/prevenção & controle , Vaccinia virus/genética , Adjuvantes Imunológicos/administração & dosagem , Animais , Apoptose , Proliferação de Células , Feminino , Humanos , Interleucina-12/administração & dosagem , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/cirurgia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/cirurgia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , Células Tumorais Cultivadas , Microambiente Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Clin Cancer Res ; 26(2): 465-476, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31767564

RESUMO

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.


Assuntos
Vacinas Anticâncer/administração & dosagem , Células-Tronco Pluripotentes Induzidas/imunologia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Experimentais/prevenção & controle , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia , Neoplasias Pancreáticas/prevenção & controle , Animais , Vacinas Anticâncer/imunologia , Chlorocebus aethiops , Progressão da Doença , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Taxa de Sobrevida , Linfócitos T/imunologia , Resultado do Tratamento
4.
Front Immunol ; 10: 2329, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31632404

RESUMO

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.


Assuntos
Doenças Transmissíveis/etiologia , Animais , Doenças Transmissíveis/diagnóstico , Doenças Transmissíveis/terapia , Cricetinae , Modelos Animais de Doenças , Suscetibilidade a Doenças , Interações Hospedeiro-Parasita , Interações Hospedeiro-Patógeno , Pesquisa
5.
Nat Commun ; 9(1): 203, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29321470

RESUMO

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.

6.
Nat Commun ; 8(1): 1395, 2017 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-29123084

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Imunoterapia/métodos , Interleucina-12/imunologia , Interleucina-12/farmacologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Neoplasias Pancreáticas/tratamento farmacológico , Adenoviridae/genética , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Cricetinae , Modelos Animais de Doenças , Feminino , Técnicas de Transferência de Genes , Humanos , Interleucina-12/efeitos adversos , Interleucina-12/química , Neoplasias Pancreáticas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Vis Exp ; (116)2016 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-27842362

RESUMO

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.


Assuntos
Sistemas CRISPR-Cas , Quebras de DNA de Cadeia Dupla , Vaccinia virus , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , RNA Guia de Cinetoplastídeos
8.
Oncoimmunology ; 4(9): e1038689, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26405610

RESUMO

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).

9.
Mol Ther Methods Clin Dev ; 2: 15035, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26417609

RESUMO

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.
Clin Cancer Res ; 21(2): 405-16, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25416195

RESUMO

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.


Assuntos
Interleucina-10/genética , Vírus Oncolíticos/genética , Neoplasias Pancreáticas/terapia , Vaccinia virus/genética , Animais , Linhagem Celular Tumoral , Linfócitos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transplante de Neoplasias , Terapia Viral Oncolítica , Neoplasias Pancreáticas/imunologia , Replicação Viral
11.
J Virol ; 87(5): 2781-90, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23269798

RESUMO

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.


Assuntos
Proteínas Proto-Oncogênicas c-akt/metabolismo , Vaccinia virus/patogenicidade , Fator A de Crescimento do Endotélio Vascular/metabolismo , Internalização do Vírus , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/virologia , Linhagem Celular Tumoral , Células Epiteliais/virologia , Genes Reporter , Humanos , Hipóxia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Interferência de RNA , RNA Interferente Pequeno , Mucosa Respiratória/virologia , Vacínia/metabolismo , Vacínia/virologia , Vaccinia virus/genética , Fator A de Crescimento do Endotélio Vascular/genética , Tropismo Viral , Replicação Viral/genética
12.
Clin Cancer Res ; 18(24): 6679-89, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23091113

RESUMO

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.


Assuntos
Neoplasias Renais/terapia , Terapia Viral Oncolítica , Neoplasias Pancreáticas/terapia , Adenoviridae/genética , Adenoviridae/imunologia , Adenoviridae/fisiologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Apoptose , Células Cultivadas , Cricetinae , Feminino , Humanos , Imunocompetência , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Mesocricetus , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Vírus Oncolíticos/fisiologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/virologia , Carga Tumoral , Vaccinia virus/genética , Vaccinia virus/imunologia , Vaccinia virus/fisiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Virus Res ; 136(1-2): 192-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18541325

RESUMO

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.


Assuntos
Genoma Viral , Vírus da Peste dos Pequenos Ruminantes/genética , RNA Viral/genética , Animais , Sequência de Bases , Côte d'Ivoire , Dados de Sequência Molecular , Nigéria , Vírus da Peste dos Pequenos Ruminantes/isolamento & purificação , Regiões Promotoras Genéticas , Ruminantes , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Proteínas Virais/genética
14.
Virus Res ; 126(1-2): 250-5, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17350130

RESUMO

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.


Assuntos
Vírus da Peste dos Pequenos Ruminantes/genética , Regiões Promotoras Genéticas , Animais , Quimera/genética , Técnicas Genéticas , Genoma Viral , Cabras , Vírus Auxiliares/genética , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/patogenicidade , Plasmídeos/genética , Vírus da Peste Bovina/genética , Ovinos , Proteínas Virais/genética
15.
Nat Chem Biol ; 2(4): 213-20, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16532013

RESUMO

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.


Assuntos
Fator de Iniciação 4A em Eucariotos/química , RNA Helicases/química , Ribossomos/química , Trifosfato de Adenosina/química , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/farmacologia , Relação Dose-Resposta a Droga , Escherichia coli/metabolismo , Fator de Iniciação 4A em Eucariotos/metabolismo , Humanos , Hidrólise , Espectroscopia de Ressonância Magnética , Modelos Genéticos , Plasmídeos/metabolismo , Poliovirus/genética , Poliovirus/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Biossíntese de Proteínas , RNA/química , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Relação Estrutura-Atividade , Transfecção
16.
J Gen Virol ; 87(Pt 4): 927-936, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16528042

RESUMO

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.


Assuntos
Enterovirus Suínos/genética , Hepacivirus/genética , Picornaviridae/genética , Picornaviridae/patogenicidade , Ribossomos/metabolismo , Regiões 5' não Traduzidas/química , Regiões 5' não Traduzidas/genética , Animais , Sequência de Bases , Códon de Iniciação , DNA Complementar/química , DNA Complementar/genética , Enterovirus Suínos/patogenicidade , Hepacivirus/patogenicidade , Humanos , Dados de Sequência Molecular , Picornaviridae/classificação , RNA Viral/química , RNA Viral/genética , Suínos/virologia
17.
J Virol ; 80(3): 1271-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16415004

RESUMO

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.


Assuntos
Enterovirus Suínos/genética , Hepacivirus/genética , RNA Viral/química , RNA Viral/genética , Animais , Sequência de Bases , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , RNA Mensageiro/química , RNA Mensageiro/genética , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie , Suínos
18.
J Virol ; 78(9): 4487-97, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15078929

RESUMO

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.


Assuntos
Hepacivirus/patogenicidade , Picornaviridae/patogenicidade , Ribossomos/metabolismo , Regiões 5' não Traduzidas , Animais , Sequência de Bases , Códon de Iniciação , Hepacivirus/genética , Dados de Sequência Molecular , Picornaviridae/genética , Biossíntese de Proteínas , RNA Viral/química , RNA Viral/metabolismo , Proteínas Ribossômicas/química , Proteínas Ribossômicas/metabolismo , Suínos/virologia
19.
J Virol ; 76(22): 11721-8, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388732

RESUMO

The teschoviruses constitute a recently defined picornavirus genus. Most of the genome sequence of the porcine teschovirus-1 (PTV) Talfan and several other strains is known. We now demonstrate that initiation of protein synthesis occurs at nucleotide (nt) 412 on the PTV Talfan RNA and that nt 1 to 405 contains an internal ribosome entry site (IRES) that functions efficiently in vitro and within mammalian cells. In comparison with other picornavirus IRES elements, the PTV IRES is relatively short and lacks a significant polypyrimidine tract near the 3' end. Expression of an enterovirus 2A protease, which induces cleavage of eIF4G within the translation initiation complex eIF4F, has little effect on the PTV IRES activity within BHK cells. The PTV IRES has a unique set of properties and represents a new class of picornavirus IRES element.


Assuntos
Regiões 5' não Traduzidas , Picornaviridae/patogenicidade , RNA Viral/química , Ribossomos/metabolismo , Suínos/virologia , Animais , Códon de Iniciação/química , Cricetinae , Cisteína Endopeptidases/metabolismo , Enterovirus/enzimologia , Picornaviridae/genética , RNA Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...