Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 3834, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34158478

RESUMO

H-1 parvovirus (H-1PV) is a promising anticancer therapy. However, in-depth understanding of its life cycle, including the host cell factors needed for infectivity and oncolysis, is lacking. This understanding may guide the rational design of combination strategies, aid development of more effective viruses, and help identify biomarkers of susceptibility to H-1PV treatment. To identify the host cell factors involved, we carry out siRNA library screening using a druggable genome library. We identify one crucial modulator of H-1PV infection: laminin γ1 (LAMC1). Using loss- and gain-of-function studies, competition experiments, and ELISA, we validate LAMC1 and laminin family members as being essential to H-1PV cell attachment and entry. H-1PV binding to laminins is dependent on their sialic acid moieties and is inhibited by heparin. We show that laminins are differentially expressed in various tumour entities, including glioblastoma. We confirm the expression pattern of laminin γ1 in glioblastoma biopsies by immunohistochemistry. We also provide evidence of a direct correlation between LAMC1 expression levels and H-1PV oncolytic activity in 59 cancer cell lines and in 3D organotypic spheroid cultures with different sensitivities to H-1PV infection. These results support the idea that tumours with elevated levels of γ1 containing laminins are more susceptible to H-1PV-based therapies.


Assuntos
Parvovirus H-1/metabolismo , Laminina/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Vírus Oncolíticos/metabolismo , Ligação Viral , Internalização do Vírus , Animais , Linhagem Celular Tumoral , Glioblastoma/patologia , Glioblastoma/terapia , Glioblastoma/virologia , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Laminina/genética , Camundongos Endogâmicos NOD , Camundongos SCID , Terapia Viral Oncolítica/métodos , Ligação Proteica , Interferência de RNA , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
2.
Cancer Gene Ther ; 22(5): 271-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25857359

RESUMO

Although H-1 parvovirus is used as an antitumor agent, not much is known about the relationship between its specific tropism and oncolytic activity. We hypothesize that VP2, a major capsid protein of H-1 virus, determines H-1-specific tropism. To assess this, we constructed chimeric H-1 viruses expressing Kilham rat virus (KRV) capsid proteins, in their complete or partial forms. Chimeric H-1 viruses (CH1, CH2 and CH3) containing the whole KRV VP2 domain could not induce cytolysis in HeLa, A549 and Panc-1 cells. However, the other chimeric H-1 viruses (CH4 and CH5) expressing a partial KRV VP2 domain induced cytolysis. Additionally, the significant cytopathic effect caused by CH4 and CH5 infection in HeLa cells resulted from preferential viral amplification via DNA replication, RNA transcription and protein synthesis. Modeling of VP2 capsid protein showed that two variable regions (VRs) (VR0 and VR2) of H-1 VP2 protein protrude outward, because of the insertion of extra amino-acid residues, as compared with those of KRV VP2 protein. This might explain the precedence of H-1 VP2 protein over KRV in determining oncolytic activity in human cancer cells. Taking these results together, we propose that the VP2 protein of oncolytic H-1 parvovirus determines its specific tropism in human cancer cells.


Assuntos
Proteínas do Capsídeo/metabolismo , Parvovirus H-1/fisiologia , Neoplasias/virologia , Infecções por Parvoviridae/virologia , Animais , Proteínas do Capsídeo/genética , Parvovirus H-1/genética , Parvovirus H-1/metabolismo , Células HeLa , Humanos , Neoplasias/genética , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Ratos , Transfecção
3.
PLoS Pathog ; 9(10): e1003671, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204256

RESUMO

Disassembly of the nuclear lamina is essential in mitosis and apoptosis requiring multiple coordinated enzymatic activities in nucleus and cytoplasm. Activation and coordination of the different activities is poorly understood and moreover complicated as some factors translocate between cytoplasm and nucleus in preparatory phases. Here we used the ability of parvoviruses to induce nuclear membrane breakdown to understand the triggers of key mitotic enzymes. Nuclear envelope disintegration was shown upon infection, microinjection but also upon their application to permeabilized cells. The latter technique also showed that nuclear envelope disintegration was independent upon soluble cytoplasmic factors. Using time-lapse microscopy, we observed that nuclear disassembly exhibited mitosis-like kinetics and occurred suddenly, implying a catastrophic event irrespective of cell- or type of parvovirus used. Analyzing the order of the processes allowed us to propose a model starting with direct binding of parvoviruses to distinct proteins of the nuclear pore causing structural rearrangement of the parvoviruses. The resulting exposure of domains comprising amphipathic helices was required for nuclear envelope disintegration, which comprised disruption of inner and outer nuclear membrane as shown by electron microscopy. Consistent with Ca⁺⁺ efflux from the lumen between inner and outer nuclear membrane we found that Ca⁺⁺ was essential for nuclear disassembly by activating PKC. PKC activation then triggered activation of cdk-2, which became further activated by caspase-3. Collectively our study shows a unique interaction of a virus with the nuclear envelope, provides evidence that a nuclear pool of executing enzymes is sufficient for nuclear disassembly in quiescent cells, and demonstrates that nuclear disassembly can be uncoupled from initial phases of mitosis.


Assuntos
Sinalização do Cálcio , Caspase 3/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Parvovirus H-1/metabolismo , Mitose , Membrana Nuclear/enzimologia , Infecções por Parvoviridae/enzimologia , Proteína Quinase C/metabolismo , Animais , Cálcio/metabolismo , Caspase 3/genética , Quinase 2 Dependente de Ciclina/genética , Parvovirus H-1/genética , Células HeLa , Humanos , Membrana Nuclear/genética , Membrana Nuclear/patologia , Membrana Nuclear/virologia , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/patologia , Proteína Quinase C/genética , Xenopus laevis
4.
J Virol ; 87(9): 5128-40, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23449783

RESUMO

The structure of single-stranded DNA (ssDNA) packaging H-1 parvovirus (H-1PV), which is being developed as an antitumor gene delivery vector, has been determined for wild-type (wt) virions and noninfectious (empty) capsids to 2.7- and 3.2-Å resolution, respectively, using X-ray crystallography. The capsid viral protein (VP) structure consists of an α-helix and an eight-stranded anti-parallel ß-barrel with large loop regions between the strands. The ß-barrel and loops form the capsid core and surface, respectively. In the wt structure, 600 nucleotides are ordered in an interior DNA binding pocket of the capsid. This accounts for ∼12% of the H-1PV genome. The wt structure is identical to the empty capsid structure, except for side chain conformation variations at the nucleotide binding pocket. Comparison of the H-1PV nucleotides to those observed in canine parvovirus and minute virus of mice, two members of the genus Parvovirus, showed both similarity in structure and analogous interactions. This observation suggests a functional role, such as in capsid stability and/or ssDNA genome recognition for encapsulation. The VP structure differs from those of other parvoviruses in surface loop regions that control receptor binding, tissue tropism, pathogenicity, and antibody recognition, including VP sequences reported to determine tumor cell tropism for oncotropic rodent parvoviruses. These structures of H-1PV provide insight into structural features that dictate capsid stabilization following genome packaging and three-dimensional information applicable for rational design of tumor-targeted recombinant gene delivery vectors.


Assuntos
Capsídeo/química , Parvovirus H-1/química , Vírion/química , Sequência de Aminoácidos , Capsídeo/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Cristalografia por Raios X , Parvovirus H-1/genética , Parvovirus H-1/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Vírion/genética , Vírion/metabolismo
5.
Clin Cancer Res ; 17(16): 5333-42, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21715567

RESUMO

PURPOSE: In previous studies, we have shown that the apathogenic rat parvovirus H-1 (H-1PV) is capable to induce regression of advanced symptomatic rat and human gliomas in a rat model, when the virus was injected in the tumor (intracranially) or intravenously. Infection with H-1PV did not provoke any pathology in nontumor tissue. This study addresses the question whether also intranasal application of this oncolytic virus is suitable and sufficient for treating gliomas in this animal model. EXPERIMENTAL DESIGN: Rat (RG-2) or human (U87) glioma cells were grafted stereotactically in the brain of rats (Wistar or RNU, respectively), and after development of tumors visible by MRI, H-1PV was instilled intranasally. Tumor regression was monitored by MRI, and survival was analyzed by Kaplan-Meier analysis. Brains from sacrificed animals were analyzed for histologic alterations, presence of viral DNA and proteins and infectious virions. In addition, distribution of virus to other organs was determined. RESULTS: A single intranasal instillation of H-1PV was sufficient to induce efficient regression of rat glioma, leading to significant prolongation of survival without any toxicity for other tissues. It is shown that the virus reaches brain and other tissues, and that the viral replication-associated (and oncolysis-associated) regulatory proteins are exclusively expressed in the tumor tissue. In rats with xenografts of human glioma, oncolytic activity of H-1PV was less pronounced, however, leading to significant prolongation of survival. CONCLUSION: In view of an ongoing clinical trial on the use of H-1PV for oncolytic virotherapy of glioma, the option of applying the virus intranasally may be a valuable alternative to invasive routes of infection.


Assuntos
Glioma/terapia , Parvovirus H-1/fisiologia , Terapia Viral Oncolítica/métodos , Infecções por Parvoviridae/terapia , Administração Intranasal , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular Tumoral , DNA Viral/análise , Modelos Animais de Doenças , Feminino , Glioma/patologia , Glioma/virologia , Parvovirus H-1/genética , Parvovirus H-1/metabolismo , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/virologia , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar , Resultado do Tratamento , Carga Tumoral , Proteínas Virais/análise , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Virol ; 84(12): 5909-22, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20375165

RESUMO

The rat parvovirus H-1 (H-1PV) attracts high attention as an anticancer agent, because it is not pathogenic for humans and has oncotropic and oncosuppressive properties. The viral nonstructural NS1 protein is thought to mediate H-1PV cytotoxicity, but its exact contribution to this process remains undefined. In this study, we analyzed the effects of the H-1PV NS1 protein on human cell proliferation and cell viability. We show that NS1 expression is sufficient to induce the accumulation of cells in G(2) phase, apoptosis via caspase 9 and 3 activation, and cell lysis. Similarly, cells infected with wild-type H-1PV arrest in G(2) phase and undergo apoptosis. Furthermore, we also show that both expression of NS1 and H-1PV infection lead to higher levels of intracellular reactive oxygen species (ROS), associated with DNA double-strand breaks. Antioxidant treatment reduces ROS levels and strongly decreases NS1- and virus-induced DNA damage, cell cycle arrest, and apoptosis, indicating that NS1-induced ROS are important mediators of H-1PV cytotoxicity.


Assuntos
Apoptose , Parvovirus H-1/metabolismo , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas não Estruturais Virais/metabolismo , Ciclo Celular , Linhagem Celular , Dano ao DNA , Parvovirus H-1/genética , Humanos , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/virologia , Proteínas não Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...