Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell Biol ; 25(5): 1669-79, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15713626

RESUMO

Alkylating agents chemically modify DNA and cause mutations that lead to cancer. In the budding yeast Saccharomyces cerevisiae, resistance to the alkylating agent methyl methanesulfonate (MMS) is mediated in part by Dap1p (damage resistance protein 1). Dap1p is related to cytochrome b5, which activates cytochrome P450 proteins, elevating the metabolism of lipids and xenobiotic compounds. We have found that Dap1p, like cytochrome b5, binds to heme and that Dap1p targets the cytochrome P450 protein Erg11p/Cyp51p. Genetic analysis indicates that Erg11p acts downstream of Dap1p. Furthermore, Dap1p regulates the stability of Erg11p, and Erg11p is stabilized in dap1Delta cells by the addition of heme. Thus, Dap1p utilizes heme to stabilize Erg11p, which in turn regulates ergosterol synthesis and MMS resistance. Dap1p homologues have been identified in numerous eukaryotes, including mammals, suggesting that the Dap1p-cytochrome P450 protein pathway is broadly conserved in eukaryotic species.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Proteínas de Membrana/fisiologia , Oxirredutases/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/metabolismo , Antineoplásicos Alquilantes/farmacologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Sistema Enzimático do Citocromo P-450/genética , Farmacorresistência Fúngica , Regulação Fúngica da Expressão Gênica , Heme/metabolismo , Proteínas Ligantes de Grupo Heme , Hemeproteínas/genética , Hemeproteínas/metabolismo , Hemeproteínas/fisiologia , Hemina/farmacologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Metanossulfonato de Metila/farmacologia , Oxirredutases/genética , Estrutura Terciária de Proteína/genética , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/enzimologia , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Deleção de Sequência/genética , Esterol 14-Desmetilase , Esteróis/metabolismo
2.
J Pharmacol Exp Ther ; 316(1): 448-55, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16234411

RESUMO

Cancer cells have varying levels of susceptibility to chemotherapeutic agents, and the proteins that direct drug susceptibility are promising targets for intervention in cancer. Hpr6 (heme-1 domain protein)/PGRMC1 (progesterone receptor membrane component 1) is overexpressed in tumors, and Hpr6 is the human homolog of a budding yeast damage resistance gene called Dap1p. Cells lacking Dap1p are damage-sensitive, and we have found that inhibition of Hpr6 expression by RNA inhibition (RNAi) increases sensitivity of breast cancer cells to chemotherapeutic drugs. Hpr6 is composed largely of a cytochrome b(5)-related heme-1 domain, and we have found that purified Hpr6 binds to heme, similar to its yeast and rodent homologues. We generated an aspartate 120-to-glycine (D120G) mutant of Hpr6 at a highly conserved site in the heme-1 domain and demonstrated that Hpr6-D120G cannot bind to heme. The Hpr6-D120G mutant was named Hpr6(hbd) for heme binding defective. We prepared an adenovirus encoding Hpr6(hbd) and found that adenovirus Hpr6(hbd) increases susceptibility of breast cancer cells to doxorubicin and camptothecin. Our findings support a model in which Hpr6, similar to its yeast homolog, binds to heme and regulates susceptibility to damaging agents.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Membrana/fisiologia , Neoplasias/tratamento farmacológico , Receptores de Progesterona/fisiologia , Antibióticos Antineoplásicos/metabolismo , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/metabolismo , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Citometria de Fluxo , Humanos , Proteínas de Membrana/metabolismo , Neoplasias/genética , Neoplasias/patologia , Plasmídeos/genética , Ligação Proteica , Interferência de RNA/efeitos dos fármacos , Receptores de Progesterona/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus/genética
3.
Tumour Biol ; 26(3): 142-6, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15970648

RESUMO

OBJECTIVE: Hpr6 (heme-1 domain protein/human progesterone receptor) is one of a family of proteins that are implicated in progesterone metabolism, resistance to genotoxic agents and steroid biosynthesis. Because these processes are frequently misregulated in tumors, we have examined the expression of Hpr6 in a group of clinical tumor samples and cancer cell lines. METHODS: Hpr6 expression was analyzed by Western blot in extracts from breast, cervix, colon and thyroid cell lines and in nonmalignant and adjacent tumor tissue from breast, colon and thyroid. Hpr6 localization was determined by immunofluorescence. RESULTS: Hpr6 expression is significantly elevated in breast tumors in comparison with matched nonmalignant tissue and demonstrated limited overexpression in colon and thyroid tumors. Hpr6 is strongly expressed in a panel of tumor cell lines originating from breast, thyroid and colon. Hpr6 localizes to the perinuclear region of the cell, consistent with a role in cell detoxification, signaling and/or sterol synthesis. CONCLUSIONS: Hpr6 homologues regulate cytochrome P450 proteins implicated in hormone, steroid and xenobiotic chemical metabolism. These are the first studies linking Hpr6 expression to cancer progression and cellular survival. Our results suggest that Hpr6 is an important marker for cancer progression and a potential anticancer therapeutic target.


Assuntos
Perfilação da Expressão Gênica , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Neoplasias/genética , Neoplasias/patologia , Receptores de Progesterona/biossíntese , Receptores de Progesterona/genética , Western Blotting , Sobrevivência Celular , Sistema Enzimático do Citocromo P-450/metabolismo , Progressão da Doença , Feminino , Humanos , Células Tumorais Cultivadas
4.
Mol Pharmacol ; 68(6): 1747-56, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16150928

RESUMO

Doxorubicin is an anthracycline antibiotic used for cancer chemotherapy. The utility of doxorubicin is limited by its inability to kill all of the cells within a tumor and by resistant cells emerging from the treated population. We have screened for genes that regulate doxorubicin susceptibility in highly tumorigenic breast cancer cells by cDNA microarray and RNA interference (RNAi) analysis, and we have identified genes associated with both proliferation and cell cycle arrest after doxorubicin treatment. We confirmed that MDA-MB-231 cells treated with doxorubicin induce the expression of carbonic anhydrase II (CAII), inhibitor of differentiation/DNA binding 2 (Id2), activating transcription factor 3 (Atf3), and the phosphatidylinositol 3-kinase 55-kDa regulatory subunit p55PIK. These genes were induced at different times and with varying specificities to different chemotherapeutic drugs. In addition to being induced at the transcriptional level, the CAII and clusterin proteins were elevated after doxorubicin treatment. CAII, Id2, p55PIK, and clusterin were not altered by doxorubicin in MCF-7 cells, a weakly tumorigenic cell line used in previous studies of doxorubicin-regulated gene expression. By inhibiting gene expression using RNAi, we found that CAII and clusterin increase cell survival after doxorubicin treatment, whereas Id2 increases susceptibility to doxorubicin. Our results support a model in which highly tumorigenic breast cancer cells induce a transcriptional response to doxorubicin that is distinct from less malignant cells. The induced genes regulate drug susceptibility positively and negatively and may be novel targets for therapeutic intervention.


Assuntos
Neoplasias da Mama/patologia , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Ciclo Celular/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Humanos , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA