Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 8(1): 15036, 2018 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-30310119

RESUMO

Lentiviral vectors are a valuable tool to deliver exogenous genes for stable expression in cells. While much progress has been made in processing lentiviral vector-containing culture medium, it remains to be explored how the production of lentiviral vector from producer cells can be increased. We initially found that co-expression of the SPRY domain-containing SOCS box protein 1 (SPSB1) promotes the production of human immunodeficiency virus type 1 (HIV-1) and lentiviral vector with increased expression of the Gag and envelope proteins and activation of the HIV-1 LTR and CMV promoter. The presence of AP-1, NF-κB and CREB/ATF recognition sites in these promoters prompted us to utilize human T-lymphotropic virus type 1 (HTLV-1) Tax for lentiviral vector production because Tax activates all these transcription factors. Co-expression of a small amount of Tax markedly increased both the expression of viral structural proteins in producer cells and release of lentiviral vector particles, resulting in a more than 10-fold enhancement of transduction efficiency. Of note, the Tax protein was not detected in the lentiviral vector particles concentrated by ultracentrifugation, supporting the safety of this preparation. Collectively, these results indicate that promoter activation in producer cells represents a promising approach to preparing high-titer lentiviral vectors.


Assuntos
Engenharia Genética , Vetores Genéticos/genética , Lentivirus/fisiologia , Regiões Promotoras Genéticas , Replicação Viral , Linhagem Celular , Produtos do Gene tax/metabolismo , Técnicas de Transferência de Genes , HIV-1/genética , Interações Hospedeiro-Patógeno , Vírus Linfotrópico T Tipo 1 Humano/genética , Humanos , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/virologia , Ligação Proteica , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Sequências Repetidas Terminais , Transdução Genética , Transfecção
2.
J Proteomics ; 75(15): 4863-73, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22687250

RESUMO

Xenotropic murine leukemia virus-related virus (XMRV) is a virus generated under artificial conditions by the recombination of 2 murine leukemia virus (MLV) proviruses, PreXMRV-1 and PreXMRV-2, during the in vivo passage of human prostate cancer cells in athymic nude mice. The molecular etiology of XMRV infection has not been characterized and its implication in human prostate cancer progression remains equivocal. As a step toward resolving this issue we developed an in vitro enzymatic assay system to characterize XMRV protease (PR)-mediated cleavage of host-cell proteins. Enzymatically-active XMRV PR protein was synthesized using a wheat-germ cell-free system. By monitoring cleavage activity of XMRV PR by AlphaScreen and 2-color immunoblot analyses, we revealed that the catalytic activity of XMRV PR is selectively blocked by the HIV PR inhibitor, Amprenavir, and identified several human tumor suppressor proteins, including PTEN and BAX, to be substrates of XMRV PR. This system may provide an attractive means for analyzing the function of retrovirus proteases and provide a technology platform for drug screening.


Assuntos
Carbamatos/química , Inibidores da Protease de HIV/química , PTEN Fosfo-Hidrolase/química , Peptídeo Hidrolases/química , Proteólise , Sulfonamidas/química , Vírus Relacionado ao Vírus Xenotrópico da Leucemia Murina/enzimologia , Proteína X Associada a bcl-2/química , Animais , Linhagem Celular Tumoral , Sistema Livre de Células/enzimologia , Furanos , Humanos , Camundongos , PTEN Fosfo-Hidrolase/metabolismo , Peptídeo Hidrolases/metabolismo , Especificidade por Substrato , Proteínas Virais , Proteína X Associada a bcl-2/metabolismo
3.
Retrovirology ; 8: 20, 2011 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-21414229

RESUMO

BACKGROUND: The involvement of xenotropic murine leukemia virus-related virus (XMRV) in prostate cancer (PC) and chronic fatigue syndrome (CFS) is disputed as its reported prevalence ranges from 0% to 25% in PC cases and from 0% to more than 80% in CFS cases. To evaluate the risk of XMRV infection during blood transfusion in Japan, we screened three populations--healthy donors (n = 500), patients with PC (n = 67), and patients with CFS (n = 100)--for antibodies against XMRV proteins in freshly collected blood samples. We also examined blood samples of viral antibody-positive patients with PC and all (both antibody-positive and antibody-negative) patients with CFS for XMRV DNA. RESULTS: Antibody screening by immunoblot analysis showed that a fraction of the cases (1.6-3.0%) possessed anti-Gag antibodies regardless of their gender or disease condition. Most of these antibodies were highly specific to XMRV Gag capsid protein, but none of the individuals in the three tested populations retained strong antibody responses to multiple XMRV proteins. In the viral antibody-positive PC patients, we occasionally detected XMRV genes in plasma and peripheral blood mononuclear cells but failed to isolate an infectious or full-length XMRV. Further, all CFS patients tested negative for XMRV DNA in peripheral blood mononuclear cells. CONCLUSION: Our data show no solid evidence of XMRV infection in any of the three populations tested, implying that there is no association between the onset of PC or CFS and XMRV infection in Japan. However, the lack of adequate human specimens as a positive control in Ab screening and the limited sample size do not allow us to draw a firm conclusion.


Assuntos
Anticorpos Antivirais/sangue , Síndrome de Fadiga Crônica/virologia , Neoplasias da Próstata/virologia , RNA Viral/sangue , Infecções por Retroviridae/complicações , Vírus Relacionado ao Vírus Xenotrópico da Leucemia Murina/isolamento & purificação , Animais , Doadores de Sangue , Linhagem Celular , Feminino , Humanos , Immunoblotting , Japão , Masculino , Camundongos , Vírus da Leucemia Murina de Moloney/imunologia , Infecções por Retroviridae/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Reação Transfusional , Vírus Relacionado ao Vírus Xenotrópico da Leucemia Murina/genética , Vírus Relacionado ao Vírus Xenotrópico da Leucemia Murina/imunologia
4.
PLoS One ; 6(1): e16121, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21264255

RESUMO

BACKGROUND: Rhesus monkey TRIM5α (TRIM5αrh) recognizes the incoming HIV-1 core through its C-terminal B30.2(PRYSPRY) domain and promotes its premature disassembly or degradation before reverse transcription. Previously, we have shown that TRIM5αrh blocks HIV-1 production through the N-terminal RBCC domain by the recognition of Gag polyproteins. Although all TRIM family proteins have RBCC domains, it remains elusive whether they possess similar late-restriction activities. METHODOLOGY/PRINCIPAL FINDINGS: We examined the antiviral spectra of TRIM5α orthologues and human TRIM family members which have a genetic locus proximal to human TRIM5α (TRIM5αhu), against primate lentiviral production. When HIV-1 virus-like particles (VLPs) were generated in the presence of TRIM5α proteins, rhesus, African green and cynomolgus monkey TRIM5α (TRIM5αag and TRIM5αcy), but not TRIM5αhu, were efficiently incorporated into VLPs, suggesting an interaction between HIV-1 Gag and TRIM5α proteins. TRIM5αrh potently restricted the viral production of HIV-1 groups M and O and HIV-2, but not simian lentiviruses including SIV(MAC)1A11, SIV(AGM)Tan-1 or SIV(AGM)SAB-1. TRIM5αhu did not show notable late restriction activities against these lentiviruses. TRIM5αag and TRIM5αcy showed intermediate restriction phenotypes against HIV-1 and HIV-2, but showed no restriction activity against SIV production. A series of chimeric TRIM5α constructs indicated that the N-terminal region of TRIM5αag and TRIM5αcy are essential for the late restriction activity, while the C-terminal region of TRIM5αcy negatively regulates the late restriction activity against HIV-1. When select human TRIM family proteins were examined, TRIM21 and 22 were efficiently incorporated into HIV-1 VLPs, while only TRIM22 reduced HIV-1 titers up to 5-fold. The antiviral activities and encapsidation efficiencies did not correlate with their relative expression levels in the producer cells. CONCLUSIONS/SIGNIFICANCE: Our results demonstrated the variations in the late restriction activities among closely related TRIM5α orthologues and a subset of human TRIM family proteins, providing further insights into the late restriction activities of TRIM proteins.


Assuntos
Antivirais/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Virais/antagonistas & inibidores , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Animais , Antivirais/farmacologia , Fatores de Restrição Antivirais , Sítios de Ligação , Proteínas de Transporte/genética , Proteínas de Transporte/farmacologia , HIV-1 , Haplorrinos , Humanos , Lentivirus , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases , Vírion/química
5.
J Biol Chem ; 285(6): 3784-3793, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19951947

RESUMO

Rhesus monkey TRIM5alpha (TRIM5alpharh) includes RING, B-box, coiled-coil, and B30.2(PRYSPRY) domains and blocks HIV-1 infection by targeting HIV-1 core through a B30.2(PRYSPRY) domain. Previously, we reported that TRIM5alpharh also blocks HIV-1 production in a B30.2(PRYSPRY)-independent manner. Efficient encapsidation of TRIM5alpharh, but not human TRIM5alpha (TRIM5alphahu), in HIV-1 virus-like particles suggests the interaction between Gag and TRIM5alpharh during viral assembly. Here, we determined responsible regions for late restriction activity of TRIM5alpharh. The RING disruption, but not the replacement with human TRIM21 RING, ablated the efficient encapsidation and the late restriction, suggesting that a RING structure was essential for the late restriction and efficient interaction with HIV-1 Gag. The prominent cytoplasmic body formation of TRIM5alpharh, which depended on the coiled-coil domain and the ensuing linker 2 region, was not required for the encapsidation. Intriguingly, TRIM5alpharh coiled-coil domain mutants (M133T and/or T146A) showed impaired late restriction activity, despite the efficient encapsidation and cytoplasmic body formation. Our results suggest that the TRIM5alpharh-mediated late restriction involves at least two distinct activities as follows: (i) interaction with HIV-1 Gag polyprotein through the N-terminal, RING, and B-box 2 regions of a TRIM5alpharh monomer, and (ii) an effector function(s) that depends upon the coiled-coil and linker 2 domains of TRIM5alpharh. We speculate that the TRIM5alpharh coiled-coil region recruits additional factor(s), such as other TRIM family proteins or a cellular protease, during the late restriction. RBCC domains of TRIM family proteins may play a role in sensing newly synthesized viral proteins as a part of innate immunity against viral infection.


Assuntos
HIV-1/fisiologia , Proteínas/fisiologia , Replicação Viral/fisiologia , Animais , Linhagem Celular , Proteínas de Fusão gag-pol/genética , Proteínas de Fusão gag-pol/metabolismo , Humanos , Immunoblotting , Macaca mulatta , Metionina/genética , Metionina/metabolismo , Mutação , Proteínas/genética , Domínios RING Finger/genética , Treonina/genética , Treonina/metabolismo , Transfecção , Ubiquitina-Proteína Ligases , Vírion/genética , Vírion/metabolismo , Montagem de Vírus , Replicação Viral/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
6.
Virology ; 397(1): 1-6, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19959199

RESUMO

The xenotropic murine leukemia virus-related virus (XMRV) is a human retrovirus, recently isolated from tissues of prostate cancer patients with impaired RNase L activity. In this study, we evaluated 10 licensed anti-HIV-1 compounds for their activity against XMRV, including protease inhibitors (PI), nucleoside reverse transcriptase (RT) inhibitors (NRTI), non-nucleoside RT inhibitors (NNRTI) and an integrase inhibitor. No PI affected XMRV production; even high concentrations of Ritonavir failed to inhibit the maturation of XMRV Gag polyproteins. Among the NRTI, NNRTI and integrase inhibitors used in this study, only AZT blocked XMRV infection and replication through inhibition of viral reverse transcription. This sensitivity of XMRV to AZT may be explained by the modest homology in the motif D sequences of HIV-1 and XMRV reverse transcriptases. If XMRV becomes established as an etiological agent for prostate cancer or other diseases, AZT may be useful for preventing or treating XMRV infections in humans.


Assuntos
Antirretrovirais/farmacologia , Retroviridae/efeitos dos fármacos , Zidovudina/farmacologia , Motivos de Aminoácidos , Humanos , Masculino , Testes de Sensibilidade Microbiana , Próstata/virologia , DNA Polimerase Dirigida por RNA/genética , Retroviridae/isolamento & purificação , Transcrição Reversa/efeitos dos fármacos , Homologia de Sequência de Aminoácidos , Proteínas Virais/genética
7.
J Virol ; 81(18): 10201-6, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17609277

RESUMO

Dominant, constitutively expressed antiretroviral factors, including TRIM5alpha and APOBEC3 proteins, are distinguished from the conventional innate immune systems and are classified as intrinsic immunity factors. Here, we demonstrate that interferon alpha (IFN-alpha) treatment upregulates TRIM5alpha mRNA in rhesus monkey cells, which correlates with the enhanced TRIM5alpha-mediated pre- and postintegration blocks of human immunodeficiency virus replication. In human cells, IFN-alpha increases the levels of TRIM5alpha mRNA, resulting in enhanced antiviral activity against N-tropic murine leukemia virus infection. These observations indicate that the TRIM5alpha-mediated antiviral effects can be orchestrated by the conventional innate immune response. It is conceivable that TRIM5alpha plays an essential role in controlling both the initial retroviral exposure and the subsequent viral dissemination in vivo.


Assuntos
Antivirais/farmacologia , Proteínas de Transporte/biossíntese , Infecções por HIV/metabolismo , HIV-1/metabolismo , Imunidade Inata/efeitos dos fármacos , Interferon-alfa/farmacologia , Proteínas/metabolismo , Replicação Viral/efeitos dos fármacos , Desaminases APOBEC , Animais , Fatores de Restrição Antivirais , Proteínas de Transporte/imunologia , Linhagem Celular , Citidina Desaminase , Citosina Desaminase/imunologia , Citosina Desaminase/metabolismo , Infecções por HIV/imunologia , HIV-1/imunologia , Humanos , Imunidade Inata/imunologia , Vírus da Leucemia Murina/imunologia , Vírus da Leucemia Murina/metabolismo , Macaca mulatta , Proteínas/imunologia , RNA Mensageiro/biossíntese , RNA Mensageiro/imunologia , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia , Replicação Viral/imunologia
8.
Mol Ther ; 15(8): 1531-6, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17505473

RESUMO

Vesicular stomatitis virus (VSV) can replicate in malignant cells more efficiently than in normal cells. Although the selective replication appears to be caused by defects in the interferon (IFN) system in malignant cells, the mechanisms which render these cells less responsive to IFN remain poorly understood. Here we present evidence that an activated RAS/Raf1/MEK/ERK pathway plays a critical role in the defects. NIH 3T3 or human primary cells stably expressing active RAS or Raf1 were rapidly killed by VSV. Although IFNalpha treatment no longer protected the RAS- or Raf1-overexpressing cells from VSV infection, responsiveness to IFNalpha was restored following treatment with the mitogen-activated protein kinase kinase (MEK) inhibitor U0126. Similarly, human cancer-derived cell lines became more responsive to IFNalpha in conjunction with U0126 treatment. Intriguingly, dual treatment with both IFNalpha and U0126 severely reduced the levels of viral RNAs in the infected cells. Moreover, cancer cells showed defects in inducing an IFNalpha-responsive factor, MxA, which is known to block VSV RNA synthesis, and U0126 restored the MxA expression. Our observations suggest that activation of the extracellular signal-regulated protein kinase (ERK) signaling leads to the defect in IFNalpha-mediated upregulation of MxA protein, which facilitates VSV oncolysis. In view of the fact that 30% of all cancers have constitutive activation of the RAS/Raf1/MEK/ERK pathway, VSV would be an ideal oncolytic virus for targeting such cancers.


Assuntos
Interferon-alfa/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Vírus da Estomatite Vesicular Indiana/metabolismo , Animais , Butadienos/farmacologia , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neoplasias/genética , Nitrilas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Vírus de RNA/genética , Vírus da Estomatite Vesicular Indiana/genética , Proteínas ras/metabolismo
9.
Nat Med ; 13(5): 631-5, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17435772

RESUMO

Mammalian cells have developed diverse strategies to restrict retroviral infection. Retroviruses have therefore evolved to counteract such restriction factors, in order to colonize their hosts. Tripartite motif-containing 5 isoform-alpha (TRIM5alpha) protein from rhesus monkey (TRIM5alpharh) restricts human immunodeficiency virus type 1 (HIV-1) infection at a postentry, preintegration stage in the viral life cycle, by recognizing the incoming capsid and promoting its premature disassembly. TRIM5alpha comprises an RBCC (RING, B-box 2 and coiled-coil motifs) domain and a B30.2(SPRY) domain. Sequences in the B30.2(SPRY) domain dictate the potency and specificity of the restriction. As TRIM5alpharh targets incoming mature HIV-1 capsid, but not precursor Gag, it was assumed that TRIM5alpharh did not affect HIV-1 production. Here we provide evidence that TRIM5alpharh, but not its human ortholog (TRIM5alphahu), blocks HIV-1 production through rapid degradation of HIV-1 Gag polyproteins. The specificity for this restriction is determined by sequences in the RBCC domain. Our observations suggest that TRIM5alpharh interacts with HIV-1 Gag during or before Gag assembly through a mechanism distinct from the well-characterized postentry restriction. This finding demonstrates a cellular factor blocking HIV-1 production by actively degrading a viral protein. Further understanding of this previously unknown restriction mechanism may reveal new targets for future anti-HIV-1 therapy.


Assuntos
Produtos do Gene gag/antagonistas & inibidores , Infecções por HIV/prevenção & controle , Proteínas/fisiologia , Replicação Viral , Animais , Linhagem Celular , Células Cultivadas , Produtos do Gene gag/metabolismo , HIV-1/fisiologia , Humanos , Rim , Macaca mulatta , Proteínas/genética , Transfecção , Ubiquitina-Proteína Ligases
10.
J Virol ; 80(15): 7769-74, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16840358

RESUMO

Murine primary cells are poorly permissive to human immunodeficiency virus type 1 (HIV-1) vector infection. Retroviral infectivity is influenced by dominant inhibitors such as TRIM5alpha. Sensitivity to TRIM5alpha is altered by interactions between cyclophilin A and the HIV-1 capsid. Here we demonstrate that competitive inhibitors of cyclophilins, cyclosporine or the related Debio-025, stimulate HIV-1 vector transduction of primary murine cells, including bone marrow and macrophages, up to 20-fold. Unexpectedly, the infectivity of an HIV-1 mutant or a simian lentivirus that does not recruit cyclophilin A is also stimulated by these drugs. We propose that cyclosporine and related compounds will be useful tools for experimental infection of murine primary cells. It is possible that HIV-1 infection of murine cells is inhibited by dominant factors related to immunophilins.


Assuntos
Ciclosporina/farmacologia , Vetores Genéticos , HIV-1/metabolismo , Imunossupressores/farmacologia , Transdução Genética , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/virologia , Ciclofilina A/farmacologia , Farmacorresistência Viral/efeitos dos fármacos , Feminino , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , HIV-1/patogenicidade , Humanos , Lentivirus/fisiologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/metabolismo , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/metabolismo , Replicação Viral/efeitos dos fármacos
11.
Biochem Biophys Res Commun ; 302(3): 489-95, 2003 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-12615060

RESUMO

Reportedly, in human immunodeficiency virus type 1 (HIV) vectors, insertion of central polypurine tract (cPPT) increased expression of transgenes for a short period. To test this for a stable condition, we constructed a series of vectors carrying a Neo(r) gene as a stable marker driven by a synthetic thymidine kinase (hTK) promoter. Transduction efficiency was increased in about 2-fold and decreased in about 8-fold by insertion of the reported 178bp and our 282bp cPPTs, respectively. PCR analyses revealed that insertion of 282bp cPPT, but not 178bp cPPT, impaired integration, although it did not deteriorate nuclear transport much. Furthermore, we found that insertion of 282bp cPPT between hTK promoter and an upstream LTR sequence reduced reporter gene activity in about 5-fold. This inhibitory effect of 282bp cPPT may partly account for the observed decrease in transduction efficiency. We suggest that actual effect of cPPT insertion should be examined in each HIV vector.


Assuntos
Técnicas Genéticas , Vetores Genéticos , HIV/genética , Purinas/química , Animais , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Proteína do Núcleo p24 do HIV/metabolismo , Células HeLa , Humanos , Lentivirus/genética , Luciferases/metabolismo , Modelos Genéticos , Plasmídeos/metabolismo , Polyomavirus/genética , Regiões Promotoras Genéticas , Timidina Quinase/genética , Fatores de Tempo , Transfecção , Transgenes
12.
Oncogene ; 21(20): 3112-20, 2002 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-12082626

RESUMO

Ini1/hsnf5 gene encodes INI1 protein, a chromatin remodeling factor associated with the SWI/SNF complex. In yeast, this complex modifies chromatin condensation to coactivate various transcriptional factors. However, in human, little is known about the SWI/SNF complex and INI1. To elucidate cellular functions of ini1, we constructed a recombinant adenovirus (AdexHA-INI1) capable of overexpressing INI1 in ini1-deficient cells. AdexHA-INI1 produced intranuclear INI1 in three ini1-deficient cell lines, changed their morphology, and decreased the proportion of viable cells. Flow cytometry and a BrdU incorporation assay showed that after the infection, growth of these cells was partially arrested at G1. In two of the three ini1-deficient cell lines, apoptosis was found to occur after the infection, as detected by the presence of cleaved poly (ADP-ribose) polymerase. To determine functional domains of INI1, we constructed plasmids expressing INI1 and its deletion mutants, which were used for a colony formation assay. Repeats 1 and 2 of INI1 were found to be required to suppress the growth of the three ini1-deficient cell lines. The results support the hypothesis that ini1 is a tumor suppressor gene and suggest a novel link between human SWI/SNF chromatin remodeling complex and apoptosis.


Assuntos
Apoptose/fisiologia , Cromatina/metabolismo , Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila , Fase G1/fisiologia , Genes Supressores de Tumor , Proteínas de Ligação a RNA , Adenoviridae/genética , Apoptose/genética , Criança , Proteínas Cromossômicas não Histona , Ensaio de Unidades Formadoras de Colônias , Replicação do DNA , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Citometria de Fluxo , Fase G1/genética , Vetores Genéticos/genética , Células HeLa/metabolismo , Células HeLa/ultraestrutura , Humanos , Plasmídeos/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Tumor Rabdoide/patologia , Rabdomiossarcoma/patologia , Ribonucleoproteína Nuclear Pequena U1/fisiologia , Proteína SMARCB1 , Deleção de Sequência , Fatores de Transcrição/fisiologia , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA