Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 146
Filtrar
1.
Viruses ; 13(2)2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33499370

RESUMO

JC Virus (JCPyV), a member of the Polyomaviridiæ family, is a human neurotropic virus with world-wide distribution. JCPyV is the established opportunistic infectious agent of progressive multifocal leukoencephalopathy, a fatal demyelinating disease, which results from the cytolytic infection of oligodendrocytes. Mutations in the regulatory region of JCPyV determine the different viral strains. Mad-1 the strain associated with PML contains two 98 base pair repeats, whereas the archetype strain (CY), which is the transmissible form of JCPyV, contains only one 98 tandem with two insertions of 62 and 23 base pairs respectively. The oncogenicity of JCPyV has been suspected since direct inoculation into the brain of rodents and primates resulted in the development of brain tumors and has been attributed to the viral protein, T-Antigen. To further understand the oncogenicity of JCPyV, a transgenic mouse colony containing the early region of the archetype strain (CY), under the regulation of its own promoter was generated. These transgenic animals developed tumors of neural crest origin, including: primitive neuroectodermal tumors, medulloblastomas, adrenal neuroblastomas, pituitary tumors, malignant peripheral nerve sheath tumors, and glioblastomas. Neoplastic cells from all different phenotypes express T-Antigen. The close parallels between the tumors developed by these transgenic animals and human CNS tumors make this animal model an excellent tool for the study of viral oncogenesis.


Assuntos
Antígenos Virais de Tumores/fisiologia , Neoplasias Encefálicas/virologia , Vírus JC/patogenicidade , Leucoencefalopatia Multifocal Progressiva/virologia , Animais , Antígenos Virais de Tumores/genética , Encéfalo/metabolismo , Neoplasias Encefálicas/patologia , Proteínas do Capsídeo/genética , Modelos Animais de Doenças , Humanos , Vírus JC/genética , Leucoencefalopatia Multifocal Progressiva/patologia , Camundongos , Camundongos Transgênicos , Proteínas Virais/genética
2.
PLoS Pathog ; 14(9): e1007276, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30188954

RESUMO

Merkel cell carcinoma (MCC) is an aggressive skin cancer with a high propensity for recurrence and metastasis. Merkel cell polyomavirus (MCPyV) is recognised as the causative factor in the majority of MCC cases. The MCPyV small tumour antigen (ST) is considered to be the main viral transforming factor, however potential mechanisms linking ST expression to the highly metastatic nature of MCC are yet to be fully elucidated. Metastasis is a complex process, with several discrete steps required for the formation of secondary tumour sites. One essential trait that underpins the ability of cancer cells to metastasise is how they interact with adjoining tumour cells and the surrounding extracellular matrix. Here we demonstrate that MCPyV ST expression disrupts the integrity of cell-cell junctions, thereby enhancing cell dissociation and implicate the cellular sheddases, A disintegrin and metalloproteinase (ADAM) 10 and 17 proteins in this process. Inhibition of ADAM 10 and 17 activity reduced MCPyV ST-induced cell dissociation and motility, attributing their function as critical to the MCPyV-induced metastatic processes. Consistent with these data, we confirm that ADAM 10 and 17 are upregulated in MCPyV-positive primary MCC tumours. These novel findings implicate cellular sheddases as key host cell factors contributing to virus-mediated cellular transformation and metastasis. Notably, ADAM protein expression may be a novel biomarker of MCC prognosis and given the current interest in cellular sheddase inhibitors for cancer therapeutics, it highlights ADAM 10 and 17 activity as a novel opportunity for targeted interventions for disseminated MCC.


Assuntos
Antígenos Virais de Tumores/fisiologia , Carcinoma de Célula de Merkel/etiologia , Poliomavírus das Células de Merkel/patogenicidade , Infecções por Polyomavirus/etiologia , Neoplasias Cutâneas/etiologia , Infecções Tumorais por Vírus/etiologia , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Carcinoma de Célula de Merkel/enzimologia , Carcinoma de Célula de Merkel/secundário , Movimento Celular , Células HEK293 , Humanos , Junções Intercelulares/patologia , Junções Intercelulares/fisiologia , Proteínas de Membrana/metabolismo , Poliomavírus das Células de Merkel/imunologia , Poliomavírus das Células de Merkel/fisiologia , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Infecções por Polyomavirus/enzimologia , Infecções por Polyomavirus/patologia , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/patologia , Infecções Tumorais por Vírus/enzimologia , Infecções Tumorais por Vírus/patologia
3.
Artigo em Inglês | MEDLINE | ID: mdl-26631824

RESUMO

INTRODUCTION: Primary cultures of human corneal epithelial (HCE) cells usually cease to grow after four or five passages. This result in a small cell yield for experiments such as the eye irritancy test represents a serious problem for human and animal corneal epithelial research. In the present study, we established an HCE cell line immortalized by simian virus 40 (SV40), a polyomavirus, and characterized the inherent morphologic and cytologic cell properties. METHODS: Primary cultured HCE cells were infected with a SV40 large T antigen (SV40 T)-expressing retrovirus, and were selected using G418 solution, an aminoglycoside antibiotic. To ensure that the immortalized cell lines express SV40 T and cytokeratin-3, a corneal epithelial-specific marker, we conducted reverse-transcription (RT)-PCR and Western blot analysis. RESULTS: These cell lines continued to grow for more than 50 generations, exhibiting a cobble stone-like appearance similar to normal HCE cells and an increased proliferation rate compared to primary cultured HCE cells. RT-PCR results showed that the immortalized cell lines expressed SV40 T while the primary cultured cells did not. In the Western blot assay, protein levels of phosphorylated (Ser15) p53 protein were significantly decreased in the immortalized cell lines while the expression of total p53 protein was constant. In addition, expression of p21(cip1), a cell cycle protein, was down-regulated in the immortalized cells. Moreover, a cornea epithelium-specific marker, cytokeratin-3 (CK-3), was expressed at equal levels in the immortalized cells and primary HCE cells. DISCUSSION: Taken together, these results indicate that immortalized HCE cell lines were successfully established using the SV40-retroviral vector. These cells may be an excellent model for detecting the adverse effects of standard toxic materials and could replace the traditional eye irritation test as an animal-free alternative method.


Assuntos
Antígenos Virais de Tumores/fisiologia , Córnea/fisiologia , Córnea/virologia , Epitélio Corneano/fisiologia , Epitélio Corneano/virologia , Vírus 40 dos Símios/fisiologia , Antígenos Virais de Tumores/administração & dosagem , Linhagem Celular Transformada , Proliferação de Células/fisiologia , Células Cultivadas , Córnea/citologia , Epitélio Corneano/citologia , Humanos
4.
J Gen Virol ; 96(12): 3532-3544, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26385761

RESUMO

Merkel cell polyomavirus (MCV) is clonally integrated in over 80 % of Merkel cell carcinomas and mediates tumour development through the expression of viral oncoproteins, the large T (LT) and small T antigens (sT). Viral integration is associated with signature mutations in the T-antigen locus that result in deletions of C-terminal replicative functions of the LT antigen. Despite these truncations, the LT LXCXE retinoblastoma (Rb) pocket protein family binding domain is retained, and the entire sT isoform is maintained intact. To investigate the ability of MCV oncoproteins to regulate host gene expression, we performed microarray analysis on cells stably expressing tumour-derived LT, tumour-derived LT along with sT, and tumour-derived LT with a mutated Rb interaction domain. Gene expression alterations in the presence of tumour-derived LT could be classified into three main groups: genes that are involved in the cell cycle (specifically the G1/S transition), genes involved in DNA replication and genes involved in cellular movement. The LXCXE mutant LT largely reversed gene expression alterations detected with the WT tumour-derived LT, while co-expression of sT did not significantly affect these patterns of gene expression. LXCXE-dependent upregulation of cyclin E and CDK2 correlated with increased proliferation in tumour-derived LT-expressing cells. Tumour-derived LT and tumour-derived LT plus sT increased expression of multiple cytokines and chemokines, which resulted in elevated levels of secreted IL-8. We concluded that, in human fibroblasts, the LXCXE motif of tumour-derived LT enhances cellular proliferation and upregulates cell cycle and immune signalling gene transcription.


Assuntos
Antígenos Virais de Tumores/fisiologia , Citocinas/metabolismo , Regulação da Expressão Gênica/imunologia , Inflamação/metabolismo , Poliomavírus das Células de Merkel/imunologia , Antígenos Virais de Tumores/imunologia , Proliferação de Células , Células Cultivadas , Citocinas/genética , Reparo do DNA , Replicação do DNA/fisiologia , Fibroblastos/fisiologia , Fibroblastos/virologia , Regulação da Expressão Gênica/fisiologia , Humanos , Transdução de Sinais/imunologia , Transcrição Gênica , Regulação para Cima
5.
J Invest Dermatol ; 135(5): 1221-1224, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25882464

RESUMO

The Merkel cell polyomavirus (MCPyV), discovered in 2008, drives the development of most Merkel cell carcinomas (MCCs) through several canonical mechanisms. A glaring gap in our knowledge remains the basis by which MCPyV, among all 12 human polyomaviruses, is the only one that causes cancer in humans. Moreover, initial attempts by numerous groups have failed to reproduce MCC in mice using oncoproteins from this polyomavirus. Verhaegen et al. report MCPyV small T-antigen-expressing transgenic mice that now provide insight into in vivo transformation mechanisms.


Assuntos
Antígenos Virais de Tumores/fisiologia , Carcinogênese/patologia , Carcinoma de Célula de Merkel/fisiopatologia , Poliomavírus das Células de Merkel/imunologia , Infecções por Polyomavirus/imunologia , Neoplasias Cutâneas/fisiopatologia , Infecções Tumorais por Vírus/imunologia , Animais
6.
Cancer Res ; 75(6): 1068-79, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25596282

RESUMO

Merkel cell polyomavirus (MCPyV) is frequently associated with Merkel cell carcinoma (MCC), a highly aggressive neuroendocrine skin cancer. Most MCC tumors contain integrated copies of the viral genome with persistent expression of the MCPyV large T (LT) and small T (ST) antigen. MCPyV isolated from MCC typically contains wild-type ST but truncated forms of LT that retain the N-terminus but delete the C-terminus and render LT incapable of supporting virus replication. To determine the oncogenic activity of MCC tumor-derived T antigens in vivo, a conditional, tissue-specific mouse model was developed. Keratin 14-mediated Cre recombinase expression induced expression of MCPyV T antigens in stratified squamous epithelial cells and Merkel cells of the skin epidermis. Mice expressing MCPyV T antigens developed hyperplasia, hyperkeratosis, and acanthosis of the skin with additional abnormalities in whisker pads, footpads, and eyes. Nearly half of the mice also developed cutaneous papillomas. Evidence for neoplastic progression within stratified epithelia included increased cellular proliferation, unscheduled DNA synthesis, increased E2F-responsive genes levels, disrupted differentiation, and presence of a DNA damage response. These results indicate that MCPyV T antigens are tumorigenic in vivo, consistent with their suspected etiologic role in human cancer.


Assuntos
Antígenos Virais de Tumores/fisiologia , Poliomavírus das Células de Merkel/imunologia , Neoplasias Cutâneas/etiologia , Animais , Cápsulas Bacterianas/fisiologia , Fator de Transcrição E2F1/fisiologia , Epitélio/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Papiloma/etiologia , Fenótipo
7.
J Invest Dermatol ; 135(5): 1415-1424, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25313532

RESUMO

Merkel cell carcinoma (MCC) is a rare and deadly neuroendocrine skin tumor frequently associated with clonal integration of a polyomavirus, Merkel cell polyomavirus (MCPyV), and MCC tumor cells express putative polyomavirus oncoprotein small T antigen (sTAg) and truncated large T antigen. Here, we show robust transforming activity of sTAg in vivo in a panel of transgenic mouse models. Epithelia of preterm sTAg-expressing embryos exhibited hyperplasia, impaired differentiation, increased proliferation, and apoptosis, and activation of a DNA damage response. Epithelial transformation did not require sTAg interaction with the protein phosphatase 2A protein complex, a tumor suppressor in some other polyomavirus transformation models, but was strictly dependent on a recently described sTAg domain that binds Fbxw7, the substrate-binding component of the Skp1/Cullin1/F-box protein ubiquitin ligase complex. Postnatal induction of sTAg using a Cre-inducible transgene also led to epithelial transformation with development of lesions resembling squamous cell carcinoma in situ and elevated expression of Fbxw7 target proteins. Our data establish that expression of MCPyV sTAg alone is sufficient for rapid neoplastic transformation in vivo, implicating sTAg as an oncogenic driver in MCC and perhaps other human malignancies. Moreover, the loss of transforming activity following mutation of the sTAg Fbxw7 binding domain identifies this domain as crucial for in vivo transformation.


Assuntos
Antígenos Virais de Tumores/fisiologia , Carcinogênese/patologia , Carcinoma de Célula de Merkel/fisiopatologia , Poliomavírus das Células de Merkel/imunologia , Infecções por Polyomavirus/imunologia , Neoplasias Cutâneas/fisiopatologia , Infecções Tumorais por Vírus/imunologia , Animais , Antígenos Virais de Tumores/imunologia , Apoptose/fisiologia , Carcinogênese/imunologia , Carcinoma de Célula de Merkel/imunologia , Carcinoma de Célula de Merkel/patologia , Proliferação de Células/fisiologia , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/patologia , Dano ao DNA/fisiologia , Modelos Animais de Doenças , Células de Merkel/imunologia , Células de Merkel/patologia , Camundongos , Camundongos Transgênicos , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia
8.
Uirusu ; 64(1): 25-34, 2014.
Artigo em Japonês | MEDLINE | ID: mdl-25765977

RESUMO

Recently, the family Polyomaviridae was classified as 3 genera, such as Orthopolyomavirus, Wukipolyomavirus which contain mammalian polyomaviruses and Avipolyomavirus which only contain avian polyomaviruses. We have recently isolated novel polyomaviruses, including Mastomys Polyoamvirus (MasPyV) and Vervet monkey Polyoamvirus-1 (VmPyV-1) by epidemiological activities and examined functions of their encoding proteins. In addition, we have been investigating the mechanisms of replication of human polyomavirus, JC polyomavirus (JCPyV). We recently obtained the results of function of JCVPyV-encoding proteins, including early protein (Large T antigen) and late proteins (VP1 and Agno). In this review, we summarized the data of our basic research activities.


Assuntos
Polyomavirus , Animais , Antígenos Virais de Tumores/fisiologia , Cafeína/farmacologia , Ciclo Celular/genética , Replicação do DNA , DNA Viral/genética , Genoma Viral/genética , Humanos , Polyomavirus/classificação , Polyomavirus/genética , Polyomavirus/fisiologia , Infecções por Polyomavirus/epidemiologia , Infecções por Polyomavirus/virologia , Infecções Tumorais por Vírus/epidemiologia , Infecções Tumorais por Vírus/virologia , Vírion/fisiologia , Replicação Viral/genética
9.
Biochem J ; 441(3): 859-67, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22023388

RESUMO

Down-regulation of Kv4.3 K⁺ channels commonly occurs in multiple diseases, but the understanding of the regulation of Kv4.3 K⁺ channels and the role of Kv4.3 K⁺ channels in pathological conditions are limited. HEK (human embryonic kidney)-293T cells are derived from HEK-293 cells which are transformed by expression of the large T-antigen. In the present study, by comparing HEK-293 and HEK-293T cells, we find that HEK-293T cells express more Kv4.3 K⁺ channels and more transcription factor Sp1 (specificity protein 1) than HEK-293 cells. Inhibition of Sp1 with Sp1 decoy oligonucleotide reduces Kv4.3 K⁺ channel expression in HEK-293T cells. Transfection of pN3-Sp1FL vector increases Sp1 protein expression and results in increased Kv4.3 K⁺ expression in HEK-293 cells. Since the ultimate determinant of the phenotype difference between HEK-293 and HEK-293T cells is the large T-antigen, we conclude that the large T-antigen up-regulates Kv4.3 K⁺ channel expression through an increase in Sp1. In both HEK-293 and HEK-293T cells, inhibition of Kv4.3 K⁺ channels with 4-AP (4-aminopyridine) or Kv4.3 small interfering RNA induces cell apoptosis and necrosis, which are completely rescued by the specific CaMKII (calcium/calmodulin-dependent protein kinase II) inhibitor KN-93, suggesting that Kv4.3 K⁺ channels contribute to cell apoptosis and necrosis through CaMKII activation. In summary, we establish: (i) the HEK-293 and HEK-293T cell model for Kv4.3 K⁺ channel study; (ii) that large T-antigen up-regulates Kv4.3 K⁺ channels through increasing Sp1 levels; and (iii) that Kv4.3 K⁺ channels contribute to cell apoptosis and necrosis through activating CaMKII. The present study provides deep insights into the mechanism of the regulation of Kv4.3 K⁺ channels and the role of Kv4.3 K⁺ channels in cell death.


Assuntos
Antígenos Virais de Tumores/fisiologia , Apoptose/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Necrose/genética , Canais de Potássio Shal/genética , Canais de Potássio Shal/fisiologia , Fator de Transcrição Sp1/fisiologia , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/metabolismo , Apoptose/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Modelos Biológicos , Necrose/metabolismo , RNA Interferente Pequeno/farmacologia , Canais de Potássio Shal/antagonistas & inibidores , Canais de Potássio Shal/metabolismo , Regulação para Cima/genética
10.
Neoplasia ; 12(9): 718-26, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20824048

RESUMO

Estrogen signaling is required for the proliferation of normal breast epithelial cells. However, prophylactic inhibition of estrogen signaling fails to prevent 56% of human breast cancer cases. The underlying mechanism is not well understood. Aberrant activation of growth factor signaling is known to provide alternative proliferation pathways in breast cells that are fully transformed, but it is not known whether activation of growth factor signaling can substitute for estrogen signaling in causing aberrant proliferation in the normal breast epithelium. Here, we report that in a retrovirus-based somatic mouse model (replication-competent ALV-LTR splice acceptor/tumor virus A) that closely mimics the evolution of sporadic human breast cancers, mammary epithelial cells harboring PyMT or activated ErbB2 evolve into tumors independent of estrogen or other ovarian functions in contrast to previous observations of estrogen-dependent cancer formation in germ line mouse models of ErbB2 activation. Importantly, ErbB2 activation in normal mammary cells causes estrogen-independent proliferation in both estrogen receptor (ER)-negative cells as well as in normally quiescent ER-positive cells. Therefore, aberrant activation of growth factor signaling contributes to estrogen-independent proliferation of both preneoplastic and cancerous mammary cells, and prophylactic therapy against both growth factor signaling and estrogen signaling may need to be considered in women with increased risk of breast cancer.


Assuntos
Antígenos Virais de Tumores/genética , Carcinoma Ductal de Mama/genética , Estrogênios/farmacologia , Genes erbB-2 , Neoplasias Mamárias Experimentais/genética , Polyomavirus/genética , Animais , Antígenos Virais de Tumores/metabolismo , Antígenos Virais de Tumores/fisiologia , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Ativação Enzimática , Feminino , Regulação Neoplásica da Expressão Gênica , Genes erbB-2/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Polyomavirus/imunologia , Polyomavirus/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-2/fisiologia , Transfecção
11.
Virology ; 406(1): 95-102, 2010 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-20673941

RESUMO

Integration of human papillomaviruses into that of the host promotes genomic instability and progression to cancer; factors that promote integration remain to be fully identified. DNA damage agents can promote double strand breaks during DNA replication providing substrates for integration and we investigated the ability of DNA damage to regulate HPV E1 and E2 mediated DNA replication. Results demonstrate that HPV E1 and E2 replication is not arrested following DNA damage, both in vivo and in vitro, while replication by SV40 Large T antigen is arrested and ATR is the candidate kinase for mediating the arrest. LTAg is a target for PIKK DNA damage signalling kinases, while E1 is not. We propose that the failure of E1 to be targeted by PIKKs allows HPV replication in the presence of DNA damaging agents. Such replication will result in double strand breaks in the viral genome ultimately promoting viral integration and cervical cancer.


Assuntos
Dano ao DNA , Replicação do DNA/genética , Replicação do DNA/fisiologia , Papillomaviridae/genética , Papillomaviridae/fisiologia , Proteínas Virais/genética , Proteínas Virais/fisiologia , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/fisiologia , Linhagem Celular , Quebras de DNA de Cadeia Dupla , DNA Viral/biossíntese , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Etoposídeo/farmacologia , Feminino , Instabilidade Genômica , Papillomavirus Humano 11/genética , Papillomavirus Humano 11/patogenicidade , Papillomavirus Humano 11/fisiologia , Humanos , Papillomaviridae/patogenicidade , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/virologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Neoplasias do Colo do Útero/etiologia , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/virologia , Integração Viral/genética , Integração Viral/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia
12.
J Virol ; 84(16): 8007-20, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20519379

RESUMO

We demonstrated previously that expression of simian virus 40 (SV40) large T antigen (LT), without a viral origin, is sufficient to induce the hallmarks of a cellular DNA damage response (DDR), such as focal accumulation of gamma-H2AX and 53BP1, via Bub1 binding. Here we expand our characterization of LT effects on the DDR. Using comet assays, we demonstrate that LT induces overt DNA damage. The Fanconi anemia pathway, associated with replication stress, becomes activated, since FancD2 accumulates in foci, and monoubiquitinated FancD2 is detected on chromatin. LT also induces a distinct set of foci of the homologous recombination repair protein Rad51 that are colocalized with Nbs1 and PML. The FancD2 and Rad51 foci require neither Bub1 nor retinoblastoma protein binding. Strikingly, wild-type LT is localized on chromatin at, or near, the Rad51/PML foci, but the LT mutant in Bub1 binding is not localized there. SV40 infection was previously shown to trigger ATM activation, which facilitates viral replication. We demonstrate that productive infection also triggers ATR-dependent Chk1 activation and that Rad51 and FancD2 colocalize with LT in viral replication centers. Using small interfering RNA (siRNA)-mediated knockdown, we demonstrate that Rad51 and, to a lesser extent, FancD2 are required for efficient viral replication in vivo, suggesting that homologous recombination is important for high-level extrachromosomal replication. Taken together, the interplay of LT with the DDR is more complex than anticipated, with individual domains of LT being connected to different subcomponents of the DDR and repair machinery.


Assuntos
Antígenos Virais de Tumores/fisiologia , Dano ao DNA , Reparo do DNA , Interações Hospedeiro-Patógeno , Vírus 40 dos Símios/patogenicidade , Fatores de Virulência/fisiologia , Animais , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Quinase 1 do Ponto de Checagem , Cromatina/química , Ensaio Cometa , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Inativação Gênica , Haplorrinos , Humanos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Proteína da Leucemia Promielocítica , Proteínas Quinases/metabolismo , Rad51 Recombinase/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
13.
J Virol ; 83(23): 12118-28, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19759150

RESUMO

Merkel cell polyomavirus (MCV) is a recently discovered human polyomavirus causing the majority of human Merkel cell carcinomas. We mapped a 71-bp minimal MCV replication core origin sufficient for initiating eukaryotic DNA replication in the presence of wild-type MCV large T protein (LT). The origin includes a poly(T)-rich tract and eight variably oriented, GAGGC-like pentanucleotide sequences (PS) that serve as LT recognition sites. Mutation analysis shows that only four of the eight PS are required for origin replication. A single point mutation in one origin PS from a naturally occurring, tumor-derived virus reduces LT assembly on the origin and eliminates viral DNA replication. Tumor-derived LT having mutations truncating either the origin-binding domain or the helicase domain also prevent LT-origin assembly. Optimal MCV replication requires coexpression of MCV small T protein (sT), together with LT. An intact DnaJ domain on the LT is required for replication but is dispensable on the sT. In contrast, PP2A targeting by sT is required for enhanced replication. The MCV origin provides a novel model for eukaryotic replication from a defined DNA element and illustrates the selective pressure within tumors to abrogate independent MCV replication.


Assuntos
Antígenos Virais de Tumores/fisiologia , Células de Merkel/virologia , Polyomavirus/fisiologia , Origem de Replicação/genética , Replicação Viral , Sequência de Bases , Sítios de Ligação , Linhagem Celular , DNA Viral/genética , DNA Viral/metabolismo , Humanos , Dados de Sequência Molecular , Mutação Puntual , Polyomavirus/genética , Ligação Proteica
14.
J Virol ; 83(18): 9521-31, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19570859

RESUMO

Simian virus 40 large T antigen (TAg) contributes to cell transformation, in part, by targeting two well-characterized tumor suppressors, pRb and p53. TAg expression affects the transcriptional circuits controlled by Rb and by p53. We have performed a microarray analysis to examine the global change in gene expression induced by wild-type TAg (TAg(wt)) and TAg mutants, in an effort to link changes in gene expression to specific transforming functions. For this analysis we have used enterocytes from the mouse small intestine expressing TAg. Expression of TAg(wt) in the mouse intestine results in hyperplasia and dysplasia. Our analysis indicates that practically all gene expression regulated by TAg in enterocytes is dependent upon its binding and inactivation of the Rb family proteins. To further dissect the role of the Rb family in the induction of intestinal hyperplasia, we have screened several lines of transgenic mice expressing a truncated TAg (TAg(N136)), which is able to interfere with the Rb pathway but lacks the functions associated with the carboxy terminus of the protein. This analysis confirmed the pivotal association between the Rb pathway and the induction of intestinal hyperplasia and revealed that upregulation of p53 target genes is not associated with the tumorigenic phenotype. Furthermore, we found that TAg(N136) was sufficient to induce intestinal hyperplasia, although the appearance of dysplasia was significantly delayed.


Assuntos
Antígenos Virais de Tumores/fisiologia , Transformação Celular Viral/genética , Fatores de Transcrição E2F/metabolismo , Enterócitos/metabolismo , Regulação da Expressão Gênica , Proteína do Retinoblastoma/metabolismo , Vírus 40 dos Símios/fisiologia , Animais , Enterócitos/virologia , Perfilação da Expressão Gênica , Hiperplasia/etiologia , Intestino Delgado/patologia , Camundongos , Camundongos Transgênicos , Proteína Supressora de Tumor p53/genética
15.
J Mater Sci Mater Med ; 20(6): 1273-80, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19267261

RESUMO

Coral has been used for bone grafts since 1970. Because coral has the advantages of good osteoconduction, biocompatibility, and biodegradation, it is also suitable for scaffolds used in bone-tissue engineering. However, the skeletons of different species of corals often vary significantly, and very few studies focus on the assessment of the permeability and mechanical properties of coral structure. In order to better understand the use of coral in bone tissue-engineering, we selected three typical models (Acropora sp., Goniopora sp., and Porites sp.) to analyze for pore size, porosity, permeability, and mechanical strength. We found Goniopora and Porites had homogenous structure and Acropora had oriented pores and irregular pore size. Acropora had the largest permeability, however, the transverse section was closed and the useful size was limited because of its habitat type. Porites had the smallest pore size and had the lowest permeability. Our data indicated that Goniopora sp. can be considered as the most promising source of scaffolds for bone-tissue engineering because of its high porosity (73%) and that its permeability and mechanics were similar to those in human cancellous bone. In conclusion, we analyzed the impact of the macroporous structure of coral on the permeability and mechanical properties that provide indicators for designing the optimal scaffold for bone-tissue engineering.


Assuntos
Osso e Ossos/metabolismo , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Antozoários , Antígenos Virais de Tumores/fisiologia , Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Fenômenos Biomecânicos , Substitutos Ósseos/química , Osso e Ossos/química , Linhagem Celular Transformada , Transformação Celular Viral , Força Compressiva , Humanos , Teste de Materiais , Microscopia Eletrônica de Varredura , Osteoblastos/citologia , Osteoblastos/ultraestrutura , Permeabilidade , Porosidade , Vírus 40 dos Símios/imunologia , Propriedades de Superfície , Resistência à Tração , Difração de Raios X
16.
Neoplasia ; 11(1): 87-95, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19107235

RESUMO

JC virus has a transforming gene encoding JC virus T-antigen (JCVT). JCVT may inactivate wild-type p53, cause chromosomal instability (CIN), and stabilize beta-catenin. A link between JCVT and CpG island methylator phenotype (CIMP) has been suggested. However, no large-scale study has examined the relations of JCVT with molecular alterations, clinical outcome, or prognosis in colon cancer. We detected JCVT expression (by immunohistochemistry) in 271 (35%) of 766 colorectal cancers. We quantified DNA methylation in eight CIMP-specific promoters (CACNA1G, CDKN2A, CRABP1, IGF2, MLH1, NEUROG1, RUNX3, and SOCS1) and eight other loci (CHFR, HIC1, IGFBP3, MGMT, MINT1, MINT31, p14, WRN) by MethyLight. We examined loss of heterozygosity in 2p, 5q, 17q, and 18q. JCVT was significantly associated with p53 expression (P < .0001), p21 loss (P < .0001), CIN (>/=2 chromosomal segments with LOH; P < .0001), nuclear beta-catenin (P = .006), LINE-1 hypomethylation (P = .002), and inversely with CIMP-high (P = .0005) and microsatellite instability (MSI) (P < .0001), but not with PIK3CA mutation. In multivariate logistic regression analysis, the associations of JCVT with p53 [adjusted odds ratio (OR), 8.45; P < .0001], CIN (adjusted OR, 2.53; P = .003), cyclin D1 (adjusted OR, 1.57; P = .02), LINE-1 hypomethylation (adjusted OR, 1.97 for a 30% decline as a unit; P = .03), BRAF mutation (adjusted OR, 2.20; P = .04), and family history of colorectal cancer (adjusted OR, 0.64; P = .04) remained statistically significant. However, JCVT was no longer significantly associated with CIMP, MSI, beta-catenin, or cyclooxygenase-2 expression in multivariate analysis. JCVT was unrelated with patient survival. In conclusion, JCVT expression in colorectal cancer is independently associated with p53 expression and CIN, which may lead to uncontrolled cell proliferation.


Assuntos
Antígenos Virais de Tumores/fisiologia , Carcinoma/genética , Instabilidade Cromossômica/fisiologia , Neoplasias Colorretais/genética , Ilhas de CpG/genética , Metilação de DNA , Genes p53 , Adulto , Idoso , Antígenos Virais de Tumores/genética , Instabilidade Cromossômica/genética , Estudos de Coortes , Metilação de DNA/fisiologia , DNA-Citosina Metilases/metabolismo , Feminino , Seguimentos , Regulação Neoplásica da Expressão Gênica , Ligação Genética , Humanos , Vírus JC/genética , Vírus JC/imunologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
17.
J Neuropathol Exp Neurol ; 67(8): 729-40, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18648329

RESUMO

The human polyomavirus JC (JCV) is a small DNA tumor virus and the etiologic agent of the progressive multifocal leukoencephalopathy. In progressive multifocal leukoencephalopathy, active JCV replication causes the lytic destruction of oligodendrocytes. The normal immune system prevents JCV replication and suppresses the virus into a state of latency so that expression of viral proteins cannot be detected. In a cellular context that is nonpermissive for viral replication, JCV can affect oncogenic transformation. For example, JCV is highly tumorigenic when inoculated into experimental animals, including rodents and monkeys. In these animal tumors, there is expression of early T-antigen but not of late capsid proteins, nor is there viral replication. Moreover, mice transgenic for JCV T-antigen alone develop tumors of neural tube origin. Detection of JCV genomic sequences and expression of viral T-antigen and agnoprotein suggest a possible association of this virus with a variety of human brain and non-CNS tumors. Here, we discuss the mechanisms involved in JCV oncogenesis, briefly review studies that do and do not support a causative role for this virus in human CNS tumors, and identify key issues for future research.


Assuntos
Neoplasias do Sistema Nervoso Central/etiologia , Neoplasias do Sistema Nervoso Central/virologia , Vírus JC/patogenicidade , Animais , Antígenos Virais de Tumores/metabolismo , Antígenos Virais de Tumores/fisiologia , Modelos Animais de Doenças , Humanos , Leucoencefalopatia Multifocal Progressiva/etiologia , Proteínas Virais Reguladoras e Acessórias/metabolismo , Proteínas Virais Reguladoras e Acessórias/fisiologia
18.
J Cell Physiol ; 215(2): 309-19, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18022798

RESUMO

The regulatory proteins of polyomaviruses, including small and large T antigens, play important roles, not only in the viral life cycle but also in virus-induced cell transformation. Unlike many other tumor viruses, the transforming proteins of polyomaviruses have no cellular homologs but rather exert their effects mostly by interacting with cellular proteins that control fundamental processes in the regulation of cell proliferation and the cell cycle. Thus, they have proven to be valuable tools to identify specific signaling pathways involved in tumor progression. Elucidation of these pathways using polyomavirus transforming proteins as tools is critically important in understanding fundamental regulatory mechanisms and hence to develop effective therapeutic strategies against cancer. In this short review, we will focus on the structural and functional features of one polyomavirus transforming protein, that is, the small t-antigen of the human neurotropic JC virus (JCV) and the simian virus, SV40.


Assuntos
Antígenos Virais de Tumores/fisiologia , Transformação Celular Viral/fisiologia , Vírus JC/fisiologia , Vírus 40 dos Símios/fisiologia , Sequência de Aminoácidos , Animais , Antígenos Virais de Tumores/genética , Humanos , Vírus JC/imunologia , Chaperonas Moleculares/fisiologia , Dados de Sequência Molecular , Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína/fisiologia , Fases de Leitura , Fase S/fisiologia , Vírus 40 dos Símios/imunologia , Serina-Treonina Quinases TOR , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
19.
Proc Natl Acad Sci U S A ; 104(48): 19011-6, 2007 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-18006659

RESUMO

Here, we show how targeting protein phosphatase 2A (PP2A), a key regulator of cellular protein phosphorylation, can either induce or prevent apoptosis depending on what other signals the cell is receiving. The oncoprotein polyoma small T interacts with PP2A to regulate survival. In the presence of growth factors, small T induces apoptosis. Akt activity, which usually promotes survival, is required for this death response, because inhibitors of Akt or PI3 kinase protect cells from death. The activation of Akt under these conditions is partial, characterized by T308 phosphorylation but not S473 phosphorylation. In the absence of growth factors, small T protects from cell death. Here, small T uses PP2A to promote phosphorylation of Akt on both T308 and S473. This effect results in a different pattern of phosphorylation of Akt substrates and shifts Akt from a proapoptotic (presence of growth factors) to an antiapoptotic mode (absence of growth factors). An intriguing possibility is that Akt phosphorylation could be therapeutically disregulated to decrease the survival of cancer cells.


Assuntos
Antígenos Virais de Tumores/fisiologia , Apoptose/fisiologia , Proteína Fosfatase 2/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Animais , Antígenos Virais de Tumores/genética , Caspase 3/análise , Meios de Cultura Livres de Soro/farmacologia , Camundongos , Células NIH 3T3/efeitos dos fármacos , Células NIH 3T3/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Fosfosserina/análise , Fosfotirosina/análise , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
20.
Biochim Biophys Acta ; 1770(8): 1145-50, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17493758

RESUMO

Receptor-activity-modifying protein (RAMP) 1 is an accessory protein of the G protein-coupled calcitonin receptor-like receptor (CLR). The CLR/RAMP1 heterodimer defines a receptor for the potent vasodilatory calcitonin gene-related peptide. A wider tissue distribution of RAMP1, as compared to that of the CLR, is consistent with additional biological functions. Here, glutathione S-transferase (GST) pull-down, coimmunoprecipitation and yeast two-hybrid experiments identified beta-tubulin as a novel RAMP1-interacting protein. GST pull-down experiments indicated interactions between the N- and C-terminal domains of RAMP1 and beta-tubulin. Yeast two-hybrid experiments confirmed the interaction between the N-terminal region of RAMP1 and beta-tubulin. Interestingly, alpha-tubulin was co-extracted with beta-tubulin in pull-down experiments and immunoprecipitation of RAMP1 coprecipitated alpha- and beta-tubulin. Confocal microscopy indicated colocalization of RAMP1 and tubulin predominantly in axon-like processes of neuronal differentiated human SH-SY5Y neuroblastoma cells. In conclusion, the findings point to biological roles of RAMP1 beyond its established interaction with G protein-coupled receptors.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas/metabolismo , Tubulina (Proteína)/metabolismo , Antígenos Virais de Tumores/fisiologia , Axônios/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Transformação Celular Viral , Clonagem Molecular , Escherichia coli/genética , Glutationa Transferase/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Microscopia Confocal , Neuroblastoma/patologia , Neurônios/metabolismo , Testes de Precipitina , Estrutura Terciária de Proteína , Proteína 1 Modificadora da Atividade de Receptores , Proteínas Modificadoras da Atividade de Receptores , Proteínas Recombinantes de Fusão/metabolismo , Vírus 40 dos Símios/fisiologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Tubulina (Proteína)/química , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...