Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Sci Rep ; 9(1): 1633, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30733501

RESUMO

Spinal muscular atrophy (SMA) is an autosomal recessive neurodegenerative disorder that causes progressive muscle weakness and is the leading genetic cause of infant mortality worldwide. SMA is caused by the loss of survival motor neuron 1 (SMN1). In humans, a nearly identical copy gene is present, called SMN2. Although SMN2 maintains the same coding sequence, this gene cannot compensate for the loss of SMN1 because of a single silent nucleotide difference in SMN2 exon 7. SMN2 primarily produces an alternatively spliced isoform lacking exon 7, which is critical for protein function. SMN2 is an important disease modifier that makes for an excellent target for therapeutic intervention because all SMA patients retain SMN2. Therefore, compounds and small molecules that can increase SMN2 exon 7 inclusion, transcription and SMN protein stability have great potential for SMA therapeutics. Previously, we performed a high throughput screen and established a class of compounds that increase SMN protein in various cellular contexts. In this study, a novel compound was identified that increased SMN protein levels in vivo and ameliorated the disease phenotype in severe and intermediate mouse models of SMA.


Assuntos
Atrofia Muscular Espinal/tratamento farmacológico , Atrofia Muscular Espinal/etiologia , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Modelos Animais de Doenças , Injeções Intraperitoneais , Camundongos Knockout , Atrofia Muscular Espinal/mortalidade , Junção Neuromuscular/efeitos dos fármacos , Índice de Gravidade de Doença , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/genética
2.
Brain Res ; 1706: 135-146, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30408476

RESUMO

We report here the finding of abnormal Golgi apparatus morphology in motor neuron like cells depleted of SMN as well as Golgi apparatus morphology in SMA patient fibroblasts. Rescue experiments demonstrate that this abnormality is dependent on SMN, but can also be rescued by expression of the COPI coatomer subunit alpha-COP. A motor neuron-like cell line containing an inducible alpha-COP shRNA was created to generate a parallel system to study knockdown of SMN or alpha-COP. Multiple assays of COPI-dependent intracellular trafficking in cells depleted of SMN demonstrate that alpha-COP function is suboptimal, including failed sequestration of plasma membrane proteins, altered binding of mRNA, and defective targeting and transport of Golgi-resident proteins.


Assuntos
Complexo I de Proteína do Envoltório/metabolismo , Complexo de Golgi/patologia , Neurônios Motores/patologia , Linhagem Celular , Proteína Coatomer/metabolismo , Citoplasma/metabolismo , Complexo de Golgi/metabolismo , Humanos , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patologia , Transporte Proteico , RNA Mensageiro/metabolismo
3.
Gene Ther ; 24(9): 520-526, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28485722

RESUMO

Spinal muscular atrophy (SMA), a prominent genetic disease of infant mortality, is caused by low levels of survival motor neuron (SMN) protein owing to deletions or mutations of the SMN1 gene. SMN2, a nearly identical copy of SMN1 present in humans, cannot compensate for the loss of SMN1 because of predominant skipping of exon 7 during pre-mRNA splicing. With the recent US Food and Drug Administration approval of nusinersen (Spinraza), the potential for correction of SMN2 exon 7 splicing as an SMA therapy has been affirmed. Nusinersen is an antisense oligonucleotide that targets intronic splicing silencer N1 (ISS-N1) discovered in 2004 at the University of Massachusetts Medical School. ISS-N1 has emerged as the model target for testing the therapeutic efficacy of antisense oligonucleotides using different chemistries as well as different mouse models of SMA. Here, we provide a historical account of events that led to the discovery of ISS-N1 and describe the impact of independent validations that raised the profile of ISS-N1 as one of the most potent antisense targets for the treatment of a genetic disease. Recent approval of nusinersen provides a much-needed boost for antisense technology that is just beginning to realize its potential. Beyond treating SMA, the ISS-N1 target offers myriad potentials for perfecting various aspects of the nucleic-acid-based technology for the amelioration of the countless number of pathological conditions.


Assuntos
Terapia Genética/métodos , Atrofia Muscular Espinal/terapia , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos/administração & dosagem , Animais , Humanos , Atrofia Muscular Espinal/genética , Estados Unidos , United States Food and Drug Administration
4.
Oncogene ; 28(27): 2492-501, 2009 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-19448667

RESUMO

The tumor suppressor p53 preserves genome integrity by inducing transcription of genes controlling growth arrest or apoptosis. Transcriptional activation involves nucleosomal perturbation by chromatin remodeling enzymes. Mammalian SWI/SNF remodeling complexes incorporate either the Brahma-related gene 1 (BRG1) or Brahma (Brm) as the ATPase subunit. The observation that tumor cell lines harboring wild-type p53 specifically maintain expression of BRG1 and that BRG1 complexes with p53 prompted us to examine the role of BRG1 in regulation of p53. Remarkably, RNAi depletion of BRG1, but not Brm, led to the activation of endogenous wild-type p53 and cell senescence. We found a proline-rich region unique to BRG1 was required for binding to the histone acetyl transferase protein, CBP, as well as to p53. Ectopic expression of a proline-rich region deletion mutant BRG1 that is defective for CBP binding inhibited p53 destabilization. Importantly, RNAi knockdown of BRG1 and CBP reduced p53 poly-ubiquitination in vivo. In support of p53 inactivation by the combined activities of BRG1 and CBP, we show that DNA damage signals promoted disassociation of BRG1 from CBP, thereby allowing p53 accumulation. Our data demonstrate a novel function of the evolutionarily conserved chromatin remodeling subunit BRG1, which cooperates with CBP to constrain p53 activity and permit cancer cell proliferation.


Assuntos
Proliferação de Células , DNA Helicases/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA , DNA Helicases/genética , Células HeLa , Humanos , Immunoblotting , Imunoprecipitação , Células Jurkat , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Nucleares/genética , Poliubiquitina/metabolismo , Ligação Proteica , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Transfecção , Proteína Supressora de Tumor p53/genética , Ubiquitinação , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
5.
Oncogene ; 26(43): 6261-8, 2007 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-17452980

RESUMO

Acetylation is thought to be a key event for p53 activation. We demonstrate that p14ARF-induced senescence of human mammary epithelial cells (MEC) is associated with p53 acetylation and requires hAda3, a component of histone acetyltransferase complexes and a p53 transcriptional coactivator. Expression of the N-terminal domain of hAda3 that binds p53 but not p300 blocked p14ARF-induced p53 acetylation and protected MECs from senescence. Consistent with these findings, the human papillomavirus 16 E6 mutant Y54D, which selectively targets hAda3 but not p53 for degradation and protects MECs from p14ARF-induced senescence, inhibited p53 acetylation. In H1299 cells, hAda3 overexpression increased p300-mediated p53 acetylation, which conversely decreased following small interfering RNA (siRNA) knockdown of hAda3. Moreover, depletion of hAda3 by siRNA inhibited endogenous p53 acetylation and accumulation of p21cip1 in response to ectopic p14ARF. These studies reveal that, in addition to its known ability to inhibit Mdm2-mediated p53 degradation, p14ARF signals through hAda3 to stimulate p53 acetylation and the induction of cell senescence.


Assuntos
Senescência Celular/fisiologia , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilação , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células Epiteliais/metabolismo , Humanos , Interferência de RNA , Telomerase/genética , Telomerase/metabolismo , Fatores de Transcrição/genética , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p53/genética
6.
Hum Mol Genet ; 10(23): 2727-36, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11726560

RESUMO

Proximal spinal muscular atrophy (SMA) is caused by mutations in the survival motor neuron gene (SMN1). In humans, two nearly identical copies of SMN exist and differ only by a single non-polymorphic C-->T nucleotide transition in exon 7. SMN1 contains a 'C' nucleotide at the +6 position of exon 7 and produces primarily full-length SMN transcripts, whereas SMN2 contains a 'T' nucleotide and produces high levels of a transcript that lacks exon 7 and a low level of full-length SMN transcripts. All SMA patients lack a functional SMN1 gene but retain at least one copy of SMN2, suggesting that the low level of full-length protein produced from SMN2 is sufficient for all cell types except motor neurons. The murine Smn gene is not duplicated or alternatively spliced. It resembles SMN1 in that the critical exon 7 +6 'C' nucleotide is conserved. We have generated Smn minigenes containing either wild-type Smn exon 7 or an altered exon 7 containing the C-->T nucleotide transition to mimic SMN2. When expressed in cultured cells or transgenic mice, the wild-type minigene produced only full-length transcripts whereas the modified minigene alternatively spliced exon 7. Furthermore, Smn exon 7 contains a critical AG-rich exonic splice enhancer sequence (ESE) analogous to the human ESE within SMN exon 7, and subtle mutations within the mESE caused a variation in Smn transcript levels. In summary, we show for the first time that the murine Smn locus can be induced to alternatively splice exon 7. These results demonstrate that SMN protein levels can be varied in the mouse by the introduction of specific mutations at the endogenous Smn locus and thereby lay the foundation for developing animals that closely 'resemble' SMA patients.


Assuntos
Processamento Alternativo , Éxons/genética , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/genética , Animais , Composição de Bases/genética , Sequência de Bases , Células COS , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Elementos Facilitadores Genéticos/genética , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação , Proteínas do Tecido Nervoso/metabolismo , Plasmídeos/genética , RNA/genética , RNA/metabolismo , Proteínas de Ligação a RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas do Complexo SMN , Homologia de Sequência do Ácido Nucleico , Proteína 1 de Sobrevivência do Neurônio Motor , Proteína 2 de Sobrevivência do Neurônio Motor , Distribuição Tecidual , Transcrição Gênica , Células Tumorais Cultivadas
7.
Gene Ther ; 8(20): 1532-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11704813

RESUMO

Spinal muscular atrophy (SMA) is a degenerative motor neuron disorder resulting from homozygous loss of the SMN1 gene. SMN2, a nearly identical copy gene, is preserved in SMA patients. A single nucleotide difference between SMN1 and SMN2 causes exon 7 skipping in the majority of SMN2 mRNA. Gene therapy through modulation of SMN2 gene transcription in SMA patients may be possible. We constructed a series of SMN mini-genes comprised of SMN exon 6 to exon 8 sequences fused to green fluorescence protein (GFP) or luciferase reporters, to monitor SMN exon 7 splicing. These reporters recapitulated the splicing patterns of the endogenous SMN gene in stable cell lines. The SMN1-luciferase reporter was approximately 3.5-fold more active than SMN2-luciferase and SMN1-GFP intensities were visually distinguishable from SMN2-GFP. We have screened chemical inducers and inhibitors of kinase pathways using stable SMN-reporter lines and found that the phosphatase inhibitor sodium vanadate specifically stimulated exon 7 inclusion within SMN2 mRNAs. This is the first compound identified that can stimulate exon 7 inclusion into transcripts derived from the endogenous SMN2 gene. These results demonstrate that this system can be utilized to identify small molecules that regulate the splicing of SMN exon 7.


Assuntos
Terapia Genética/métodos , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/terapia , Proteínas do Tecido Nervoso/genética , Splicing de RNA/efeitos dos fármacos , Vanadatos/farmacologia , Fusão Gênica Artificial/métodos , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Inibidores Enzimáticos/farmacologia , Éxons , Expressão Gênica , Proteínas de Fluorescência Verde , Humanos , Luciferases/genética , Proteínas Luminescentes/genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Fosforilação , Proteínas de Ligação a RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas do Complexo SMN , Proteína 1 de Sobrevivência do Neurônio Motor , Proteína 2 de Sobrevivência do Neurônio Motor , Transfecção/métodos
8.
Mol Cell Biol ; 21(17): 5913-24, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11486030

RESUMO

We have reported that the papillomavirus E2 protein binds the nuclear factor AMF1 (also called G-protein pathway suppressor 2 or GPS2) and that their interaction is necessary for transcriptional activation by E2. It has also been shown that AMF1 can influence the activity of cellular transcription factors. These observations led us to test whether AMF1 regulates the functions of p53, a critical transcriptional activator that integrates stress signals and regulates cell cycle and programmed cell death. We report that AMF1 associates with p53 in vivo and in vitro and facilitates the p53 response by augmenting p53-dependent transcription. Overexpression of AMF1 in U2OS cells increases basal level p21(WAF1/CIP1) expression and causes a G(1) arrest. U2OS cells stably overexpressing AMF1 show increased apoptosis upon exposure to UV irradiation. These data demonstrate that AMF1 modulates p53 activities.


Assuntos
Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Linhagem Celular , Linhagem Celular Transformada , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Fase G1 , Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Nucleares/genética , Ligação Proteica , Proteínas Repressoras/genética , Spodoptera , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Raios Ultravioleta
9.
Biochemistry ; 40(5): 1293-9, 2001 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-11170455

RESUMO

E6AP is a cellular protein that binds cancer-related papillomaviral E6 proteins. The E6 binding domain, called E6ap, is located on an 18-amino acid segment of E6AP. The corresponding peptide was synthesized and its structure determined by nuclear magnetic resonance spectroscopy. The overall structure of the peptide is helical. A consensus E6-binding sequence among different E6 interacting proteins contains three conserved hydrophobic residues. In the structure of the E6AP peptide, the three conserved leucines (Leu 9, Leu 12, and Leu 13) form a hydrophobic patch on one face of the alpha-helix. Substitution of any of these leucines with alanine abolished binding to E6 protein, indicating that the entire hydrophobic patch is necessary. Mutation of a glutamate to proline, but not alanine, also disrupted the interaction between E6 and E6AP protein, suggesting that the E6-binding motif of the E6AP protein must be helical when bound to E6. Comparison of the E6ap structure and mutational results with those of another E6-binding protein (E6BP/ERC-55) indicates the existence of a general E6-binding motif.


Assuntos
Ligases/química , Ligases/genética , Proteínas Oncogênicas Virais/metabolismo , Proteínas Repressoras , Sequência de Aminoácidos , Papillomavirus Bovino 1/química , Papillomavirus Bovino 1/genética , Dicroísmo Circular , Cristalografia por Raios X , Análise Mutacional de DNA , Humanos , Ligases/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ressonância Magnética Nuclear Biomolecular , Papillomaviridae/química , Papillomaviridae/genética , Ligação Proteica/genética , Soluções , Ubiquitina-Proteína Ligases
10.
Hum Mol Genet ; 9(19): 2869-77, 2000 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-11092763

RESUMO

Spinal muscular atrophy (SMA) is caused by mutations in the SMN (survival of motor neurons) gene and there is a correlation between disease severity and levels of functional SMN protein. Studies of structure-function relationships in SMN protein may lead to a better understanding of SMA pathogenesis. Self-association of the spinal muscular atrophy protein, SMN, is important for its function in RNA splicing. Biomolecular interaction analysis core analysis now shows that SMN self-association occurs via SMN regions encoded by exons 2b and 6, that exon 2b encodes a binding site for SMN-interacting protein-1 and that interaction occurs between exon 2- and 4-encoded regions within the SMN monomer. The presence of two separate self-association sites suggests a novel mechanism by which linear oligomers or closed rings might be formed from SMN monomers.


Assuntos
Éxons/genética , Proteínas de Homeodomínio/metabolismo , Atrofia Muscular Espinal/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Proteínas Repressoras/metabolismo , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Ligação Competitiva , Técnicas Biossensoriais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Dimerização , Mapeamento de Epitopos , Humanos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Fragmentos de Peptídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA , Proteínas Recombinantes de Fusão/metabolismo , Proteínas do Complexo SMN , Homeobox 2 de Ligação a E-box com Dedos de Zinco
11.
Proc Natl Acad Sci U S A ; 97(17): 9618-23, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10931943

RESUMO

Spinal muscular atrophy (SMA), a common motor neuron disease in humans, results from loss of functional survival motor neuron (SMN1) alleles. A nearly identical copy of the gene, SMN2, fails to provide protection from SMA because of a single translationally silent nucleotide difference in exon 7. This likely disrupts an exonic splicing enhancer and causes exon 7 skipping, leading to abundant production of a shorter isoform, SMN2Delta7. The truncated transcript encodes a less stable protein with reduced self-oligomerization activity that fails to compensate for the loss of SMN1. This report describes the identification of an in vivo regulator of SMN mRNA processing. Htra2-beta1, an SR-like splicing factor and ortholog of Drosophila melanogaster transformer-2, promoted the inclusion of SMN exon 7, which would stimulate full-length SMN2 expression. Htra2-beta1 specifically functioned through and bound an AG-rich exonic splicing enhancer in SMN exon 7. This effect is not species-specific as expression of Htra2-beta1 in human or mouse cells carrying an SMN2 minigene dramatically increased production of full-length SMN2. This demonstrates that SMN2 mRNA processing can be modulated in vivo. Because all SMA patients retain at least one SMN2 copy, these results show that an in vivo modulation of SMN RNA processing could serve as a therapeutic strategy to prevent SMA.


Assuntos
Éxons/genética , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/metabolismo , Splicing de RNA/genética , Animais , Sequência de Bases , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Terapia Genética , Humanos , Camundongos , Dados de Sequência Molecular , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Proteínas do Complexo SMN , Fatores de Processamento de Serina-Arginina , Proteína 1 de Sobrevivência do Neurônio Motor , Proteína 2 de Sobrevivência do Neurônio Motor
12.
J Biol Chem ; 275(40): 30894-900, 2000 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-10887172

RESUMO

Bovine papillomavirus type 1 (BPV-1) is a small DNA virus that causes fibropapillomas of the host. BPV-1 has served as the prototype for studies of the molecular biology of the papillomaviruses. BPV-1 efficiently induces anchorage-independent growth and focus formation in murine C127 cells. The transforming properties of BPV-1 primarily reside in two genes, E5 and E6. Each of these genes is sufficient to transform cells. Although no independent transformation activity has been detected for E7, it was shown to be required for full transformation of C127 by BPV-1. We investigated the biological activities of BPV-1 E7 in several assays. Our results indicate that expression of BPV-1 E7 sensitizes cells to tumor necrosis factor alpha (TNF)-induced apoptosis. The TNF-induced apoptosis in E7-expressing cells was accompanied by increased release of arachidonic acid, indicating that phospholipase A(2) was activated. Unlike the E7 proteins from the cancer-related human papillomaviruses, the BPV-1 E7 protein does not associate efficiently with the retinoblastoma protein (pRB) in vitro, nor does it significantly affect the pRB levels in cultured cells. Furthermore, BPV-1 E7 sensitizes Rb-null cells to TNF-induced apoptosis. These studies indicate that BPV-1 E7 can sensitize cells to apoptosis through mechanisms that are independent of pRB.


Assuntos
Apoptose , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/metabolismo , Proteína do Retinoblastoma/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Ácido Araquidônico/metabolismo , Western Blotting , Morte Celular , Divisão Celular , Linhagem Celular , Separação Celular , Corantes/farmacologia , Cicloeximida/farmacologia , Ativação Enzimática , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Formazans/farmacologia , Glutationa Transferase/metabolismo , Humanos , Vírus da Leucemia Murina/genética , Camundongos , Proteínas Oncogênicas Virais/genética , Fosfolipases A/metabolismo , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sais de Tetrazólio/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
13.
J Virol ; 74(13): 5872-9, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10846067

RESUMO

The cellular protein AMF-1 (Gps2) positively modulates gene expression by the papillomavirus E2 protein (D. E. Breiding et al., Mol. Cell. Biol. 17:7208-7219, 1997). We show here that AMF-1 also binds the transcriptional coactivator p300 in vitro and in vivo. E2 interacted weakly with p300. These observations led to a model in which AMF-1 recruits p300 into a complex with E2. Cotransfection of AMF-1 or p300 stimulated levels of E2-dependent transcription, while cotransfection of both AMF-1 and p300 showed an additive effect. The functional significance of p300 recruitment for E2 transactivation was evidenced by repression of E2-activated transcription by adenovirus E1A, which inhibits both coactivator and acetylase activities of p300. Antibodies to AMF-1 or E2 immunoprecipitated histone acetylase activity from cell lysates. Western blotting using antibody against acetyl-lysine failed to detect acetylation of AMF-1 or E2 in complex with p300. These results suggest that AMF-1 facilitates the recruitment of p300 and its histone acetylase activity into complexes with E2 and represents a novel mechanism of transcriptional activation.


Assuntos
Papillomavirus Bovino 1 , Proteínas de Ligação a DNA/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Proteínas Repressoras , Transativadores/metabolismo , Proteínas Virais/metabolismo , Acetilação , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Animais , Sítios de Ligação , Bovinos , Linhagem Celular , Precipitação Química , Proteínas de Ligação a DNA/genética , Proteínas Fúngicas/genética , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Nucleares/genética , Proteínas Oncogênicas Virais/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Spodoptera/citologia , Transativadores/genética , Ativação Transcricional , Células Tumorais Cultivadas , Proteínas Virais/genética
14.
Hum Mol Genet ; 9(2): 259-65, 2000 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-10607836

RESUMO

The survival motor neuron genes, SMN1 and SMN2, encode identical proteins; however, only homo- zygous loss of SMN1 correlates with the development of spinal muscular atrophy (SMA). We have previously shown that a single non-polymorphic nucleotide difference in SMN exon 7 dramatically affects SMN mRNA processing. SMN1 primarily produces a full-length RNA whereas SMN2 expresses dramatically reduced full-length RNA and abundant levels of an aberrantly spliced transcript lacking exon 7. The importance of proper exon 7 processing has been underscored by the identification of several mutations within splice sites adjacent to exon 7. Here we show that an AG-rich exonic splice enhancer (ESE) in the center of SMN exon 7 is required for inclusion of exon 7. This region functioned as an ESE in a heterologous context, supporting efficient in vitro splicing of the Drosophila double-sex gene. Finally, the protein encoded by the exon-skipping event, Delta7, was less stable than full-length SMN, providing additional evidence of why SMN2 fails to compensate for the loss of SMN1 and leads to the development of SMA.


Assuntos
Elementos Facilitadores Genéticos/genética , Éxons/genética , Atrofia Muscular Espinal/genética , Proteínas do Tecido Nervoso/genética , Processamento Alternativo/genética , Sequência de Bases , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Humanos , Dados de Sequência Molecular , Proteínas de Ligação a RNA , Proteínas do Complexo SMN , Proteína 1 de Sobrevivência do Neurônio Motor , Proteína 2 de Sobrevivência do Neurônio Motor , Células Tumorais Cultivadas
15.
Biochemistry ; 38(49): 16115-24, 1999 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-10587434

RESUMO

Papillomaviral E2 proteins participate in viral DNA replication and transcriptional regulation. We have solved the solution structure of the DNA-binding domain of the E2 protein from bovine papillomavirus (BPV-1). The structure calculation used 2222 distance and 158 dihedral angle restraints for the homodimer (202 residues in total), which were derived from homonuclear and heteronuclear multidimensional nuclear magnetic resonance (NMR) spectroscopic data. The root-mean-square deviation for structured regions of the monomer when superimposed to the average is 0.73 +/- 0.10 A for backbone atoms and 1.42 +/- 0.16 A for heavy atoms. The 101 residue construct used in this study (residues 310-410) is about 4.5 kcal/mol more stable than a minimal domain comprising the C-terminal 85 amino acid residues (residues 326-410). The structure of the core domain contained within BPV-1 E2 is similar to the corresponding regions of other papilloma viral E2 proteins. Here, however, the extra N-terminal 16 residues form a flap that covers a cavity at the dimer interface and play a role in DNA binding. Interactions between residues in the N-terminal extension and the core domain correlate with the greater stability of the longer form of the protein relative to the minimal domain.


Assuntos
Papillomavirus Bovino 1/química , DNA Viral/metabolismo , Proteínas de Ligação a DNA/química , Fragmentos de Peptídeos/química , Proteínas Virais/química , Animais , Bovinos , Cristalografia por Raios X , DNA Viral/química , Proteínas de Ligação a DNA/metabolismo , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Fragmentos de Peptídeos/metabolismo , Desnaturação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Soluções , Relação Estrutura-Atividade , Termodinâmica , Ureia , Proteínas Virais/metabolismo
16.
Virus Res ; 65(2): 141-54, 1999 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-10581387

RESUMO

Papillomavirus E1 and E2 proteins co-operation in viral DNA replication is mediated by protein-protein interactions that lead to formation of an E1-E2 complex. To identify the domains involved, portions of the two proteins were expressed as fusions to the DNA-binding protein LexA or the transactivation domain of VP16 and analyzed by the yeast two-hybrid system. The C-terminal 266 amino acids of BPV1 E1 (E1C266) interacted strongly with E2 in the yeast system and in a mammalian two-hybrid assay. VP16-E1C266 interacted with a region encompassing amino acids 1-200 of the transactivation domain of E2 that was fused to LexA. The interaction between E1 full length and E2 was clearly observed only when E1 was expressed as LexA-E1 chimera. In addition, we found that in the LexA context also the N-terminal region encompassing the first 340 amino acids of E1 (E1N340) interacted with E2 full length. The interactions of E1N340 and E1C266 with E2 were confirmed also by in vitro binding studies. These observations demonstrate that two distinct regions of E1 mediate the interaction with E2 in vivo.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas Virais/química , Animais , Papillomavirus Bovino 1/química , Papillomavirus Bovino 1/fisiologia , Bovinos , DNA Helicases/química , DNA Viral/metabolismo , Elementos Facilitadores Genéticos/fisiologia , Transativadores/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Replicação Viral/fisiologia
17.
J Virol ; 73(9): 7297-307, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10438818

RESUMO

The E6 proteins from cervical cancer-associated human papillomavirus (HPV) types such as HPV type 16 (HPV-16) induce proteolysis of the p53 tumor suppressor protein through interaction with E6-AP. We have previously shown that human mammary epithelial cells (MECs) immortalized by HPV-16 E6 display low levels of p53. HPV-16 E6 as well as other cancer-related papillomavirus E6 proteins also binds the cellular protein E6BP (ERC-55). To explore the potential functional significance of these interactions, we created and analyzed a series of E6 mutants for their ability to interact with E6-AP, p53, and E6BP in vitro. While there was a similar pattern of binding among these E6 targets, a subset of mutants differentiated E6-AP binding, p53 binding, and p53 degradation activities. These results demonstrated that E6 binding to E6-AP is not sufficient for binding to p53 and that E6 binding to p53 is not sufficient for inducing p53 degradation. The in vivo activity of these HPV-16 E6 mutants was tested in MECs. In agreement with the in vitro results, most of these p53 degradation-defective E6 mutants were unable to reduce the p53 level in early-passage MECs. Interestingly, several mutants that showed severely reduced ability for interacting with E6-AP, p53, and E6BP in vitro efficiently immortalized MECs. These immortalized cells exhibited low p53 levels at late passage. Furthermore, mutants defective for p53 degradation but able to immortalize MECs were also identified, and the immortal cells retained normal levels of p53 protein. These results imply that multiple functions of HPV-16 E6 contribute to MEC immortalization.


Assuntos
Mama/virologia , Proteínas Oncogênicas Virais/fisiologia , Papillomaviridae/fisiologia , Proteínas Repressoras , Animais , Mama/citologia , Transformação Celular Viral , Células Epiteliais , Feminino , Humanos , Mutagênese Sítio-Dirigida , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Coelhos , Proteína Supressora de Tumor p53/metabolismo
18.
J Biol Chem ; 274(35): 24819-27, 1999 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-10455154

RESUMO

Human papillomavirus type 16 (HPV-16) E6 has been shown to prevent or enhance apoptosis depending on the stimulus and cell type. Here we present evidence that HPV-16 E6 sensitized murine fibrosarcoma L929 cells to tumor necrosis factor alpha (TNF)-induced cytolysis. The E6-enhanced cytolysis correlated with a precedent increase in reactive oxygen species (ROS) level and antioxidant treatment could completely block the E6-dependent sensitization. These findings represent the first demonstration of a link between a viral oncogene-sensitized cytolysis and ROS. Previous studies have shown conflicting results regarding whether TNF-induced cytolysis of L929 cells is through necrosis or apoptosis. Here we report that, although L929 cells underwent DNA fragmentation after exposure to TNF, they retained the morphology of intact nuclei while gaining permeability to propidium iodide, features characteristic of necrosis rather than apoptosis. We confirmed that the broad spectrum caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone markedly increased the susceptibility of L929 cells to TNF, and further demonstrated that E6 enhanced this susceptibility, which again correlated with increased ROS accumulation. We showed that the expression of E6 in L929 cells did not alter the stability of p53, and the cells retained a p53 response to actinomycin D. Furthermore, two E6 mutants defective for p53 degradation in other systems exhibited differential effects on TNF sensitization. These results suggest that the enhancement of TNF-induced L929 cytolysis by E6 is independent of p53 degradation. We also found that TNF-induced activation of NF-kappaB did not account for the enhanced TNF susceptibility by E6.


Assuntos
Proteínas Oncogênicas Virais/metabolismo , Proteínas Repressoras , Fator de Necrose Tumoral alfa/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citometria de Fluxo , Genes Reporter , Humanos , Células Jurkat , Camundongos , Microscopia de Fluorescência , NF-kappa B/genética , Necrose , Proteínas Oncogênicas Virais/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
19.
Hum Mol Genet ; 8(7): 1177-83, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10369862

RESUMO

Spinal muscular atrophy (SMA) is a recessive disorder characterized by loss of motor neurons in the spinal cord. It is caused by mutations in the telomeric survival motor neuron 1 ( SMN1 ) gene. Alterations within an almost identical copy gene, the centromeric survival motor neuron 2 ( SMN2 ) gene produce no known phenotypic effect. The exons of the two genes differ by just two nucleotides, neither of which alters the encoded amino acids. At the genomic level, only five nucleotides that differentiate the two genes from one another have been reported. The entire genomic sequence of the two genes has not been determined. Thus, differences which might explain why SMN1 is the SMA gene are not readily apparent. In this study, we have completely sequenced and compared genomic clones containing the SMN genes. The two genes show striking similarity, with the homology being unprecedented between two different yet functional genes. The only critical difference in an approximately 32 kb region between the two SMN genes is the C->T base change 6 bp inside exon 7. This alteration but not other variations in the SMN genes affects the splicing pattern of the genes. The majority of the transcript from the SMN1 locus is full length, whereas the majority of the transcript produced by the SMN2 locus lacks exon 7. We suggest that the exon 7 nucleotide change affects the activity of an exon splice enhancer. In SMA patients, the loss of SMN1 but the presence of SMN2 results in low levels of full-length SMN transcript and therefore low SMN protein levels which causes SMA.


Assuntos
Processamento Alternativo , Atrofia Muscular Espinal/genética , Proteínas do Tecido Nervoso/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Genótipo , Humanos , Dados de Sequência Molecular , Mutação Puntual , Proteínas de Ligação a RNA , Proteínas do Complexo SMN , Análise de Sequência de DNA , Proteína 1 de Sobrevivência do Neurônio Motor , Proteína 2 de Sobrevivência do Neurônio Motor
20.
Hum Mol Genet ; 8(7): 1219-26, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10369867

RESUMO

Spinal muscular atrophy (SMA) is an inherited neuro-muscular disease characterized by specific degeneration of spinal cord anterior horn cells and subsequent muscle atrophy. Survival motor neuron ( SMN ), located on chromosome 5q13, is the SMA-determining gene. In the nucleus, SMN is present in large foci called gems, the function of which is not yet known, while cytoplasmic SMN has been implicated in snRNP biogenesis. In SMA patients, SMN protein levels and the number of gems generally correlate with disease severity, suggesting a critical nuclear function for SMN. In a screen for proteins associated with the nuclear transcription activator 'E2' of papillomavirus, two independent SMN cDNAs were isolated. The E2 and SMN proteins were found to associate specifically in vitro and in vivo. Expression of SMN enhanced E2-dependent transcriptional activation, and patient-derived SMN missense mutations reduced E2 gene expression. Our results demonstrate that SMN interacts with a nuclear transcription factor and imply that SMN may serve a role in regulating gene expression. These observations suggest that SMA may in part result from abnormal gene expression and that E2 may influence viral gene expression through SMN interaction.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Atrofia Muscular Espinal/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transativadores/metabolismo , Proteínas Virais/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Escherichia coli , Regulação da Expressão Gênica , Células HeLa , Humanos , Atrofia Muscular Espinal/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/isolamento & purificação , Ligação Proteica , Proteínas de Ligação a RNA , Proteínas do Complexo SMN , Saccharomyces cerevisiae
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA