Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Genet Couns ; 25(1): 18-24, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26354339

RESUMO

The Jewish community has traditionally taken ownership of its health, and has taken great strides to raise awareness about genetic issues that affect the community, such as Tay-Sachs disease and Hereditary Breast and Ovarian Cancer syndrome. Thanks in part to these heightened awareness efforts, many Orthodox Jewish individuals are now using genetics services as they begin to plan their families. Due to unique cultural and religious beliefs and perceptions, the Orthodox Jewish patients who seek genetic counseling face many barriers to a successful counseling session, and often seek the guidance of programs such as the Program for Jewish Genetic Health (PJGH). In this article, we present clinical vignettes from the PJGH's clinical affiliate, the Reproductive Genetics practice at the Montefiore Medical Center. These cases highlight unique features of contemporary premarital counseling and screening within the Orthodox Jewish Community, including concerns surrounding stigma, disclosure, "marriageability," the use of reproductive technologies, and the desire to include a third party in decision making. Our vignettes demonstrate the importance of culturally-sensitive counseling. We provide strategies and points to consider when addressing the challenges of pre- and post-test counseling as it relates to genetic testing in this population.


Assuntos
Aconselhamento/métodos , Aconselhamento Genético/métodos , Comportamentos Relacionados com a Saúde/etnologia , Judaísmo , Casamento/etnologia , Educação Pré-Natal/métodos , Tomada de Decisões , Feminino , Predisposição Genética para Doença/prevenção & controle , Testes Genéticos , Humanos , Masculino , Valores Sociais/etnologia
2.
JIMD Rep ; 16: 31-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24850235

RESUMO

Mutations in glucosidase, beta, acid (GBA) are associated with cognitive impairment in Parkinson disease (PD) as well as dementia with Lewy bodies. For both of these diseases, dementia and hallucinations are typically treated with cholinesterase inhibitors and antipsychotics. However, in some lysosomal storage disorders certain antipsychotic medications are poorly tolerated. This study examined cholinesterase inhibitor and antipsychotic use in monoallelic GBA-related PD to explore potential pharmacogenetic relationships. Monoallelic GBA mutation carriers with PD (GBA-PD) with at least two clinic visits (n = 34) were matched for age-of-onset and gender to GBA and leucine-rich repeat kinase 2 (LRRK2) mutation negative idiopathic PD subjects (IPD) (n = 60). Information regarding cholinesterase inhibitor and antipsychotic use as well as impaired cognition (UPDRS Mentation >1) and hallucinations (UPDRS Thought Disorder >1) were obtained. GBA-PD more frequently reported hallucinations (HR = 5.0; p = 0.01) and they were more likely to have cognitive impairment but this was not statistically significant (HR 2.2, p = 0.07). Antipsychotic use was not significantly different between GBA-PD and IPD (HR = 1.9; p = 0.28), but GBA-PD were more likely to have sustained cholinesterase inhibitor use (HR = 3.1; p = 0.008), even after adjustment for cognition and hallucinations. Consistent with reports of worse cognition, GBA-PD patients are more likely to use cholinesterase inhibitors compared to IPD. While there was no difference in antipsychotic use between IPD and GBA-PD, persistent use of quetiapine in GBA-PD suggests that it is tolerated and that a significant interaction is unlikely. Further prospective study in larger samples with more extensive cognitive assessment is warranted to better understand pharmacogenetic relationships in GBA-PD.

3.
Parkinsonism Relat Disord ; 19(2): 186-91, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23062841

RESUMO

BACKGROUND: Heterozygous glucocerebrosidase (GBA) mutations are the leading genetic risk factor for Parkinson disease, yet imaging correlates, particularly transcranial sonography, have not been extensively described. METHODS: To determine whether GBA mutation heterozygotes with Parkinson disease demonstrate hyperechogenicity of the substantia nigra, transcranial sonography was performed in Ashkenazi Jewish Parkinson disease subjects, tested for the eight most common Gaucher disease mutations and the LRRK2 G2019S mutation, and in controls. [(18)F]-fluorodeoxyglucose or [(18)F]-fluorodopa positron emission tomography is also reported from a subset of Parkinson disease subjects with heterozygous GBA mutations. RESULTS: Parkinson disease subjects with heterozygous GBA mutations (n = 23) had a greater median maximal area of substantia nigral echogenicity compared to controls (n = 34, aSNmax = 0.30 vs. 0.18, p = 0.007). There was no difference in median maximal area of nigral echogenicity between Parkinson disease groups defined by GBA and LRRK2 genotype: GBA heterozygotes; GBA homozygotes/compound heterozygotes (n = 4, aSNmax = 0.27); subjects without LRRK2 or GBA mutations (n = 32, aSNmax = 0.27); LRRK2 heterozygotes/homozygotes without GBA mutations (n = 27, aSNmax = 0.28); and GBA heterozygotes/LRRK2 heterozygotes (n = 4, aSNmax = 0.32, overall p = 0.63). In secondary analyses among Parkinson disease subjects with GBA mutations, maximal area of nigral echogenicity did not differ based on GBA mutation severity or mutation number. [(18)F]-fluorodeoxyglucose (n = 3) and [(18)F]-fluorodopa (n = 2) positron emission tomography in Parkinson disease subjects with heterozygous GBA mutations was consistent with findings in idiopathic Parkinson disease. CONCLUSIONS: Both transcranial sonography and positron emission tomography are abnormal in GBA mutation associated Parkinson disease, similar to other Parkinson disease subjects.


Assuntos
Glucosilceramidase/genética , Doença de Parkinson/diagnóstico por imagem , Doença de Parkinson/genética , Idoso , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos , Ultrassonografia Doppler Transcraniana
4.
Curr Top Microbiol Immunol ; 302: 205-34, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16620030

RESUMO

The past two decades of gene targeting experiments have allowed us to make significant strides towards understanding how the Myc/Max/Mad network influences multiple aspects of cellular behavior during development. Here we summarize the findings obtained from the myc/max/mad knockout mice generated to date, namely those in which the N-myc, c-myc, L-myc, mad1, mxi1, mad3, mnt, or max genes have been targeted. A compilation of lessons we have learned from these myc/max/mad knockout mouse models, and suggestions as to where future efforts could be focused, are also presented.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/deficiência , Proteínas Proto-Oncogênicas c-myc/deficiência , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Desenvolvimento Embrionário , Feminino , Genes myc , Humanos , Camundongos , Camundongos Knockout , Morfogênese , Neoplasias/etiologia , Fenótipo , Gravidez , Proteínas Proto-Oncogênicas c-myc/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia
5.
Biochemistry ; 40(11): 3354-62, 2001 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-11258956

RESUMO

The c-Myc oncoprotein contributes to oncogenesis by activating and repressing a repertoire of genes involved in cellular proliferation, metabolism, and apoptosis. Increasing evidence suggests that the repressor function of c-Myc is critical for transformation. Therefore, identifying and characterizing Myc-repressed genes is imperative to understanding the mechanisms of Myc-induced tumorigenesis. Here, we employ NIH 3T3 cell lines harboring c-Myc-ER or N-Myc-ER to dissect the relationship between Myc activation and caveolin-1 expression. In this well-established inducible system, treatment with estrogen like molecules, such as tamoxifen, leads to activation of Myc, but in a tightly controlled fashion. Using this approach, we show that Myc activation induces the repression of caveolin-1 expression at the transcriptional level. We also provide two independent lines of evidence suggesting that caveolin-1 is a direct target of Myc: (i) the effect of Myc activation on caveolin-1 expression is independent of new protein synthesis, as revealed through the use of cycloheximide; and (ii) Myc-mediated repression of the caveolin-1 promoter is dependent on an intact INR sequence. Moreover, we show that expression of caveolin-1, via an adenoviral vector approach, can suppress cell transformation that is mediated by Myc activation. In support of these observations, treatment with an adenoviral vector harboring anti-sense caveolin-1 specifically potentiates transformation induced by Myc activation. Taken together, our results indicate that caveolin-1 is a direct target of Myc repression, and they also provide evidence for an additional mechanism by which Myc repression can elicit a malignant phenotype.


Assuntos
Caveolinas/antagonistas & inibidores , Caveolinas/genética , Regulação da Expressão Gênica/genética , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/fisiologia , Proteínas Repressoras/fisiologia , Transcrição Gênica , Células 3T3 , Regiões 5' não Traduzidas/antagonistas & inibidores , Regiões 5' não Traduzidas/genética , Animais , Caveolina 1 , Caveolinas/biossíntese , Caveolinas/fisiologia , Transformação Celular Neoplásica/genética , Regulação para Baixo/genética , Camundongos , Fenótipo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Receptores de Estrogênio/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transfecção
6.
Mol Cell Biol ; 21(5): 1810-8, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238917

RESUMO

Cells isolated from individuals with Cockayne syndrome (CS) have a defect in transcription-coupled DNA repair, which rapidly corrects certain DNA lesions located on the transcribed strand of active genes. Despite this DNA repair defect, individuals with CS group A (CSA) or group B (CSB) do not exhibit an increased spontaneous or UV-induced cancer rate. In order to investigate the effect of CSB deficiency on spontaneous carcinogenesis, we crossed CSB(-/-) mice with cancer-prone mice lacking the p16(Ink4a)/p19(ARF) tumor suppressor locus. CSB(-/-) mice are sensitive to UV-induced skin cancer but show no increased rate of spontaneous cancer. CSB(-/-) Ink4a/ARF(-/-) mice developed 60% fewer tumors than Ink4a/ARF(-/-) animals and demonstrated a longer tumor-free latency time (260 versus 150 days). Moreover, CSB(-/-) Ink4a/ARF(-/-) mouse embryo fibroblasts (MEFs) exhibited a lower colony formation rate after low-density seeding, a lower rate of H-Ras-induced transformation, slower proliferation, and a lower mRNA synthesis rate than Ink4a/ARF(-/-) MEFs. CSB(-/-) Ink4a/ARF(-/-) MEFs were also more sensitive to UV-induced p53 induction and UV-induced apoptosis than were Ink4a/ARF(-/-) MEFs. In order to investigate whether the apparent antineoplastic effect of CSB gene disruption was caused by sensitization to genotoxin-induced (p53-mediated) apoptosis or by p53-independent sequelae, we also generated p53(-/-) and CSB(-/-) p53(-/-) MEFs. The CSB(-/-) p53(-/-) MEFs demonstrated lower colony formation efficiency, a lower proliferation rate, a lower mRNA synthesis rate, and a higher rate of UV-induced cell death than p53(-/-) MEFs. Collectively, these results indicate that the antineoplastic effect of CSB gene disruption is at least partially p53 independent; it may result from impaired transcription or from apoptosis secondary to environmental or endogenous DNA damage.


Assuntos
Síndrome de Cockayne/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , DNA Helicases/genética , DNA Helicases/fisiologia , Neoplasias/genética , Proteínas/genética , Fatores Etários , Animais , Apoptose , Divisão Celular , Cruzamentos Genéticos , Reparo do DNA , Enzimas Reparadoras do DNA , Fibroblastos/metabolismo , Fibrossarcoma/metabolismo , Citometria de Fluxo , Genes p53/genética , Predisposição Genética para Doença , Genótipo , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Linfoma/metabolismo , Camundongos , Camundongos Knockout , Proteínas de Ligação a Poli-ADP-Ribose , RNA Mensageiro/metabolismo , Fatores de Tempo , Transformação Genética , Proteína Supressora de Tumor p14ARF , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta , Proteínas ras/metabolismo
7.
Nat Genet ; 24(2): 113-9, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10655054

RESUMO

Myc and Mad family proteins regulate multiple biological processes through their capacity to influence gene expression directly. Here we show that the basic regions of Myc and Mad proteins are not functionally equivalent in oncogenesis, have separable E-box-binding activities and engage both common and distinct gene targets. Our data support the view that the opposing biological actions of Myc and Mxi1 extend beyond reciprocal regulation of common gene targets. Identification of differentially regulated gene targets provides a framework for understanding the mechanism through which the Myc superfamily governs the growth, proliferation and survival of normal and neoplastic cells.


Assuntos
Transformação Celular Neoplásica , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Repressoras , Fatores de Transcrição/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Apoptose , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Divisão Celular , Linhagem Celular , Sobrevivência Celular , Sequência Conservada , Proteínas de Ligação a DNA/química , Sequências Hélice-Alça-Hélice , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Proteínas Proto-Oncogênicas c-myc/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/química , Transfecção , Proteínas Supressoras de Tumor
8.
Science ; 287(5456): 1253-8, 2000 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-10678830

RESUMO

Accelerated telomere loss has been proposed to be a factor leading to end-stage organ failure in chronic diseases of high cellular turnover such as liver cirrhosis. To test this hypothesis directly, telomerase-deficient mice, null for the essential telomerase RNA (mTR) gene, were subjected to genetic, surgical, and chemical ablation of the liver. Telomere dysfunction was associated with defects in liver regeneration and accelerated the development of liver cirrhosis in response to chronic liver injury. Adenoviral delivery of mTR into the livers of mTR(-/-) mice with short dysfunctional telomeres restored telomerase activity and telomere function, alleviated cirrhotic pathology, and improved liver function. These studies indicate that telomere dysfunction contributes to chronic diseases of continual cellular loss-replacement and encourage the evaluation of "telomerase therapy" for such diseases.


Assuntos
Terapia Genética , Cirrose Hepática Experimental/terapia , Regeneração Hepática , Fígado/patologia , Telomerase/genética , Adenoviridae/genética , Animais , Apoptose , Tetracloreto de Carbono/toxicidade , Técnicas de Transferência de Genes , Vetores Genéticos , Hepatectomia , Fígado/enzimologia , Cirrose Hepática Experimental/enzimologia , Cirrose Hepática Experimental/patologia , Cirrose Hepática Experimental/fisiopatologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mitose , Baço/enzimologia , Telomerase/metabolismo , Telômero/fisiologia , Telômero/ultraestrutura , Fator de Crescimento Transformador beta/metabolismo
9.
Oncogene ; 18(38): 5349-55, 1999 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-10498888

RESUMO

The pathogenetic basis of prostate cancer remains highly elusive; its clarification could be facilitated greatly by laboratory and clinical models of the disease. Although the genetically manipulated mouse has been invaluable for the modeling of other human cancer types, it has fared less well with respect to prostate cancer. Nevertheless, several highly valuable transgenic models exist and are highlighted in this review. Emerging reagents and strategies may allow us to use the mouse more effectively to define the molecular, cellular and physiological events that lead to prostate cancer initiation and progression.


Assuntos
Modelos Animais de Doenças , Neoplasias da Próstata/genética , Animais , Humanos , Masculino , Camundongos , Camundongos Transgênicos
10.
Bioessays ; 20(10): 808-18, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9819568

RESUMO

The functions of Myc in transformation and transactivation are countered by the suppressive actions of the Mad(Mxi1) family. Mad(Mxi1) proteins not only compete with Myc for dimerization to Max and binding to Myc/Max consensus sites but also recruit powerful repressors of gene expression. A prediction of the yin-yang relationship between Myc and Mad(Mxi1) families would be that the latter constitutes a new class of tumor suppressors. Here, we review the current literature on the Mad(Mxi1) family, with particular attention paid to the molecular mechanisms by which these proteins antagonize the actions of Myc in normal and neoplastic cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Proteínas Repressoras/fisiologia , Proteínas de Saccharomyces cerevisiae , Fatores de Transcrição/fisiologia , Ativação Transcricional/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Fatores de Transcrição de Zíper de Leucina Básica , Sítios de Ligação , Ligação Competitiva , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Cromatina/genética , Sequência Consenso , Dimerização , Células Eucarióticas/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Histona Desacetilases , Camundongos , Camundongos Knockout , Modelos Genéticos , Família Multigênica , Neoplasias/genética , Proteínas Nucleares/fisiologia , Correpressor 1 de Receptor Nuclear , Proteínas Supressoras de Tumor
11.
Nature ; 393(6684): 483-7, 1998 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-9624006

RESUMO

Mxi1 belongs to the Mad (Mxi1) family of proteins, which function as potent antagonists of Myc oncoproteins. This antagonism relates partly to their ability to compete with Myc for the protein Max and for consensus DNA binding sites and to recruit transcriptional co-repressors. Mad(Mxi1) proteins have been suggested to be essential in cellular growth control and/or in the induction and maintenance of the differentiated state. Consistent with these roles, mxi1 may be the tumour-suppressor gene that resides at region 24-26 of the long arm of chromosome 10. This region is a cancer hotspot, and mutations here may be involved in several cancers, including prostate adenocarcinoma. Here we show that mice lacking Mxi1 exhibit progressive, multisystem abnormalities. These mice also show increased susceptibility to tumorigenesis either following carcinogen treatment or when also deficient in Ink4a. This cancer-prone phenotype may correlate with the enhanced ability of several mxi1-deficient cell types, including prostatic epithelium, to proliferate. Our results show that Mxi1 is involved in the homeostasis of differentiated organ systems, acts as a tumour suppressor in vivo, and engages the Myc network in a functionally relevant manner.


Assuntos
Envelhecimento/fisiologia , Proteínas de Ligação a DNA/fisiologia , Neoplasias/etiologia , Fatores de Transcrição/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Divisão Celular , Transformação Celular Neoplásica , Genes Supressores de Tumor , Rim/crescimento & desenvolvimento , Rim/patologia , Masculino , Camundongos , Especificidade de Órgãos , Fenótipo , Próstata/crescimento & desenvolvimento , Próstata/patologia , Proteínas Supressoras de Tumor
12.
Cell ; 92(6): 713-23, 1998 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-9529248

RESUMO

The INK4a gene encodes two distinct growth inhibitors--the cyclin-dependent kinase inhibitor p16Ink4a, which is a component of the Rb pathway, and the tumor suppressor p19Arf, which has been functionally linked to p53. Here we show that p19Arf potently suppresses oncogenic transformation in primary cells and that this function is abrogated when p53 is neutralized by viral oncoproteins and dominant-negative mutants but not by the p53 antagonist MDM2. This finding, coupled with the observations that p19Arf and MDM2 physically interact and that p19Rrf blocks MDM2-induced p53 degradation and transactivational silencing, suggests that p19Arf functions mechanistically to prevent MDM2's neutralization of p53. Together, our findings ascribe INK4a's potent tumor suppressor activity to the cooperative actions of its two protein products and their relation to the two central growth control pathways, Rb and p53.


Assuntos
Genes p16/fisiologia , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/genética , Biotina , Linhagem Celular/química , Linhagem Celular/citologia , Linhagem Celular/fisiologia , Fragmentação do DNA , Nucleotídeos de Desoxiuracil , Cristalino/citologia , Camundongos , Proteínas de Neoplasias/genética , Osteoblastos/química , Osteoblastos/citologia , Osteoblastos/fisiologia , Proteínas Proto-Oncogênicas c-mdm2 , Proteína do Retinoblastoma/metabolismo , Coloração e Rotulagem , Transformação Genética , Proteína Supressora de Tumor p14ARF
13.
J Biol Chem ; 273(7): 3803-7, 1998 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-9461560

RESUMO

In this study, we investigated the molecular basis of the ability of staurosporine to induce G1 arrest in murine embryonic fibroblasts (MEFs). We used MEFs from transgenic mice lacking several negative regulators of the G1/S phase transition including cells from mice lacking p53, p21, retinoblastoma (Rb), or p16 genes. We found that p53 function was not essential for staurosporine-induced G1 arrest. In contrast, MEFs from mice lacking Rb genes showed approximately a 70% reduced capacity to arrest in the G1 phase following staurosporine treatment. In support of a role for Rb in staurosporine-induced G1 arrest, rat embryonic fibroblasts stably overexpressing cyclin D1/Cdk4(R24C) exhibited approximately a 50% reduced G1 arrest response to staurosporine. The role of Rb in determining the degree of staurosporine-induced G1 arrest did not depend on the function of the cyclin-dependent kinase inhibitors p16 or p21 because MEFs lacking either of these genes were still capable of undergoing G1 arrest following staurosporine exposure. Our studies provide evidence of an important role for the Rb protein in determining the degree of staurosporine-induced G1 arrest in the first cell cycle.


Assuntos
Fase G1/fisiologia , Proteína do Retinoblastoma/fisiologia , Estaurosporina/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Ciclina D1/metabolismo , Fibroblastos , Citometria de Fluxo , Camundongos , Camundongos Transgênicos , Supressão Genética/genética
14.
Proc Natl Acad Sci U S A ; 95(4): 1392-7, 1998 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-9465025

RESUMO

Proper control of the mammalian cell cycle requires the function of cyclin-dependent kinase (CDK) inhibitors. The p21 family currently includes three distinct genes, p21, p27(Kip1), and p57(Kip2), that share a common N-terminal domain for binding to and inhibiting the kinase activity of CDK-cyclin complexes. The p21 protein also binds to proliferating cell nuclear antigen (PCNA) through a separate C-terminal domain affecting DNA replication and repair. The p27 and p57 proteins also each contain unique C-terminal domains whose functions are unknown. Here we show that the human p57 protein, like p21, contains a PCNA-binding domain within its C terminus that, when separated from its N-terminal CDK-cyclin binding domain, can prevent DNA replication in vitro and S phase entry in vivo. Disruption of either CDK/cyclin or PCNA binding partially reduced p57's ability to suppress myc/RAS-mediated transformation in primary cells, while loss of both inhibitory functions completely eliminated p57's suppressive activity. Thus, control of cell cycle and suppression of cell transformation by p57 require both CDK and PCNA inhibitory activity, and disruption of either or both functions may lead to uncontrolled cell growth.


Assuntos
Proteínas de Ciclo Celular , Transformação Celular Neoplásica/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/metabolismo , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Supressoras de Tumor , Sequência de Aminoácidos , Animais , Síndrome de Beckwith-Wiedemann/genética , Sítios de Ligação , Ciclo Celular , Cromossomos Humanos Par 11 , Inibidor de Quinase Dependente de Ciclina p27 , Inibidor de Quinase Dependente de Ciclina p57 , DNA/biossíntese , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Dados de Sequência Molecular , Ligação Proteica , Ratos , Relação Estrutura-Atividade
15.
Nature ; 387(6628): 49-55, 1997 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9139821

RESUMO

Normal mammalian growth and development are highly dependent on the regulation of the expression and activity of the Myc family of transcription factors. Mxi1-mediated inhibition of Myc activities requires interaction with mammalian Sin3A or Sin3B proteins, which have been purported to act as scaffolds for additional co-repressor factors. The identification of two such Sin3-associated factors, the nuclear receptor co-repressor (N-CoR) and histone deacetylase (HD1), provides a basis for Mxi1/Sin3-induced transcriptional repression and tumour suppression.


Assuntos
Regulação da Expressão Gênica , Histona Desacetilases/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Proteínas de Saccharomyces cerevisiae , Fatores de Transcrição/fisiologia , Células 3T3 , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proteínas de Ligação a DNA/fisiologia , Proteínas Fúngicas/fisiologia , Genes myc , Humanos , Camundongos , Correpressor 1 de Receptor Nuclear , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/genética , Transcrição Gênica , Proteínas Supressoras de Tumor
16.
Proc Natl Acad Sci U S A ; 94(4): 1235-40, 1997 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-9037036

RESUMO

Biochemical and biological activities of Myc oncoproteins are highly dependent upon their association with another basic region helix-loop-helix/leucine zipper (bHLH/LZ) protein, Max. Our previous observation that the DNA-binding/dimerization region of Max is absolutely conserved throughout vertebrate evolution provided the basis for a yeast two-hybrid interaction screen that led to the isolation of the Drosophila Myc (dMyc1) protein. Structural conservation in regions of known functional significance is consistent with the ability of dMyc1 to interact with vertebrate Max, to transactivate gene expression in yeast cells, and to cooperate with activated H-RAS to effect the malignant transformation of primary mammalian cells. The ability of P-element-mediated ectopic expression of dmyc1 to reverse a subset of the phenotypic alterations associated with the diminutive mutation suggests that diminutive may correspond to dmyc1. This finding, along with the localization of dmyc1 expression to zones of high proliferative activity in the embryo, implicates dMyc1 as an integral regulator of Drosophila growth and development.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA/metabolismo , Drosophila/genética , Genes myc , Proteínas Proto-Oncogênicas c-myc/metabolismo , Sequência de Aminoácidos , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Fatores de Transcrição de Zíper de Leucina Básica , Diferenciação Celular , Células Cultivadas , Drosophila/crescimento & desenvolvimento , Fibroblastos/citologia , Sequências Hélice-Alça-Hélice , Mitose , Dados de Sequência Molecular , Fenótipo , Ligação Proteica , Ratos , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Fatores de Transcrição/metabolismo , Ativação Transcricional
18.
J Investig Dermatol Symp Proc ; 1(2): 128-35, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9627706

RESUMO

Myc family oncoproteins function as sequence-specific transcription factors that are believed to regulate the expression of genes governing cellular growth, differentiation, and programmed cell death. Activities of Myc are countered by those of Mad and Mxi1, two related members of the Myc superfamily. Mad and Mxi1 compete with Myc for common elements and interact with putative transcriptional repressors. While the precise role of the Myc superfamily in cutaneous biology remains to be determined, findings from a number of organ systems suggest that the regulated expression and function of its members are intimately correlated with proper development and physiology. Reviewed here are current data on Myc superfamily function with references where relevant to cutaneous processes with the ultimate goal of providing a framework upon which these proteins can be exploited in gene therapeutic approaches for diseases of the skin, including neoplasia.


Assuntos
Proteínas Proto-Oncogênicas c-myc/fisiologia , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Pele/crescimento & desenvolvimento , Animais , Diferenciação Celular , Divisão Celular , Terapia Genética , Histona Desacetilases , Humanos , Neoplasias/etiologia , Dermatopatias/terapia , Fatores de Transcrição/fisiologia
19.
Oncogene ; 12(5): 1165-72, 1996 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-8649810

RESUMO

Mxi1 is a basic region helix-loop-helix leucine zipper (bHLH/LZ) protein that, in association with Max, antagonizes Myc oncogenic activities. A possible mechanistic basis for Mxi1-mediated repression was provided by the recent demonstration that the repressive potential of Mxi1 correlates with its ability to physically associate with mSin3B, one of two mammalian homologues of the yeast transcriptional repressor SIN3. Here, we sought to characterize more fully the physical properties of the second homologue, mSin3A and to determine whether the recruitment of mSin3A by Mxi1 is indeed required for anti-Myc activity. Transient transfection of mammalian cells showed that the mSin3A protein can associate with the strong repressive isoform of Mxi1 (Mxi1-SR) and that, like other Myc superfamily members, both mSin3A and Mxi1-SR localize to the nucleus. From a developmental standpoint, a comparative analysis of Myc, Mxi1-SR and Sin3A expression during postnatal mouse development and in differentiating mouse erythroleukemia (MEL) cells revealed that dramatic and reciprocal changes in Myc and Mxi1-SR mRNA levels are accompanied by minimal stage-specific changes in mSin3A gene expression. This constant expression profile, coupled with the observation that over-expression of mSin3A does not augment the anti-Myc activity of Mxi1-SR in the rat embryo fibroblast (REF) transformation assay, suggests that mSin3A is not a limiting factor in the regulation of Myc superfamily function. Finally, a mSin3A-Mxi1 fusion protein, in which the amino terminal mSin3-interacting domain of Mxi1-SR was replaced with the full-length mSin3A, exhibited a level of repression activity equivalent to, or greater than, the level of repression obtained with Mxi1-SR. Taken together, these observations directly demonstrate that the amino-terminal repression domain of Mxi1-SR functions solely to recruit mSin3A and possibly other proteins like mSin3A and this association is necessary for the anti-Myc activity of Mxi1-SR.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Sequências Hélice-Alça-Hélice , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Diferenciação Celular , Núcleo Celular/metabolismo , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/química , Leucemia Eritroblástica Aguda , Camundongos , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , RNA Mensageiro/metabolismo , Ratos , Proteínas Repressoras/química , Complexo Correpressor Histona Desacetilase e Sin3 , Fatores de Transcrição/química , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor
20.
Proc Natl Acad Sci U S A ; 92(18): 8488-92, 1995 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-7667316

RESUMO

The positive effects of Myc on cellular growth and gene expression are antagonized by activities of another member of the Myc superfamily, Mad. Characterization of the mouse homolog of human mad on the structural level revealed that domains shown previously to be required in the human protein for anti-Myc repression, sequence-specific DNA-binding activity, and dimerization with its partner Max are highly conserved. Conservation is also evident on the biological level in that both human and mouse mad can antagonize the ability of c-myc to cooperate with ras in the malignant transformation of cultured cells. An analysis of c-myc and mad gene expression in the developing mouse showed contrasting patterns with respect to tissue distribution and developmental stage. Regional differences in expression were more striking on the cellular level, particularly in the mouse and human gastrointestinal system, wherein c-Myc protein was readily detected in immature proliferating cells at the base of the colonic crypts, while Mad protein distribution was restricted to the postmitotic differentiated cells in the apex of the crypts. An increasing gradient of Mad was also evident in the more differentiated subcorneal layers of the stratified squamous epithelium of the skin. Together, these observations support the view that both downregulation of Myc and accumulation of Mad may be necessary for progression of precursor cells to a growth-arrested, terminally differentiated state.


Assuntos
Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Proteínas Repressoras , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/genética , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas c-myc/genética , Ratos , Pele/metabolismo , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA