Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Leukemia ; 27(4): 813-22, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23138183

RESUMO

The t(10;11)(p12;q23) translocation and the t(10;11)(p12;q14) translocation, which encode the MLL (mixed lineage leukemia)-AF10 and CALM (clathrin assembly lymphoid myeloid leukemia)-AF10 fusion oncoproteins, respectively, are two recurrent chromosomal rearrangements observed in patients with acute myeloid leukemia and acute lymphoblastic leukemia. Here, we demonstrate that MLL-AF10 and CALM-AF10-mediated transformation is dependent on the H3K79 methyltransferase Dot1l using genetic and pharmacological approaches in mouse models. Targeted disruption of Dot1l using a conditional knockout mouse model abolished in vitro transformation of murine bone marrow cells and in vivo initiation and maintenance of MLL-AF10 or CALM-AF10 leukemia. The treatment of MLL-AF10 and CALM-AF10 transformed cells with EPZ004777, a specific small-molecule inhibitor of Dot1l, suppressed expression of leukemogenic genes such as Hoxa cluster genes and Meis1, and selectively impaired proliferation of MLL-AF10 and CALM-AF10 transformed cells. Pretreatment with EPZ004777 profoundly decreased the in vivo spleen-colony-forming ability of MLL-AF10 or CALM-AF10 transformed bone marrow cells. These results show that patients with leukemia-bearing chromosomal translocations that involve the AF10 gene may benefit from small-molecule therapeutics that inhibit H3K79 methylation.


Assuntos
Inibidores Enzimáticos/farmacologia , Inativação Gênica , Metiltransferases/antagonistas & inibidores , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Fusão Oncogênica/genética , Animais , Apoptose , Western Blotting , Ciclo Celular , Proliferação de Células , Imunofluorescência , Humanos , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Oncogene ; 32(8): 939-46, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23160372

RESUMO

The human protein methyltransferases (PMTs) constitute a large enzyme class composed of two families, the protein lysine methyltransferases (PKMTs) and the protein arginine methyltransferases (PRMTs). Examples have been reported of both PKMTs and PRMTs that are genetically altered in specific human cancers, and in several cases these alterations have been demonstrated to confer a unique dependence of the cancer cells on PMT enzymatic activity for the tumorigenic phenotype. Examples of such driver alterations in PMTs will be presented together with a review of current efforts towards the discovery and development of small-molecule inhibitors of these enzymes as personalized cancer therapeutics.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Medicina de Precisão/métodos , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/genética , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Epigenômica , Humanos , Terapia de Alvo Molecular/métodos , Neoplasias/genética , Proteínas Metiltransferases/metabolismo
3.
Proc Natl Acad Sci U S A ; 104(19): 8071-6, 2007 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-17470784

RESUMO

Histone deacetylase inhibitors (HDACi) can elicit a range of biological responses that affect tumor growth and survival, including inhibition of cell cycle progression, induction of tumor cell-selective apoptosis, suppression of angiogenesis, and modulation of immune responses, and show promising activity against hematological malignancies in clinical trials. Using the Emu-myc model of B cell lymphoma, we screened tumors with defined genetic alterations in apoptotic pathways for therapeutic responsiveness to the HDACi vorinostat. We demonstrated a direct correlation between induction of tumor cell apoptosis in vivo and therapeutic efficacy. Vorinostat did not require p53 activity or a functional death receptor pathway to kill Emu-myc lymphomas and mediate a therapeutic response but depended on activation of the intrinsic apoptotic pathway with the proapoptotic BH3-only proteins Bid and Bim playing an important role. Our studies provide important information regarding the mechanisms of action of HDACi that have broad implications regarding stratification of patients receiving HDACi therapy alone or in combination with other anticancer agents.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/uso terapêutico , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/uso terapêutico , Linfoma de Células B/tratamento farmacológico , Animais , Proteínas Reguladoras de Apoptose/fisiologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/fisiologia , Proteína 11 Semelhante a Bcl-2 , Genes myc , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Vorinostat , Proteína bcl-X/fisiologia
4.
Proc Natl Acad Sci U S A ; 101(5): 1241-6, 2004 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-14734806

RESUMO

Histone deacetylase (HDAC) inhibitors (HDACi) cause cancer cell growth arrest and/or apoptosis in vivo and in vitro. The HDACi suberoylanilide hydroxamic acid (SAHA) is in phase I/II clinical trials showing significant anticancer activity. Despite wide distribution of HDACs in chromatin, SAHA alters the expression of few genes in transformed cells. p21(WAF1) is one of the most commonly induced. SAHA does not alter the expression of p27(KIPI), an actively transcribed gene, or globin, a silent gene, in ARP-1 cells. Here we studied SAHA-induced changes in the p21(WAF1) promoter of ARP-1 cells to better understand the mechanism of HDACi gene activation. Within 1 h, SAHA caused modifications in acetylation and methylation of core histones and increased DNase I sensitivity and restriction enzyme accessibility in the p21(WAF1) promoter. These changes did not occur in the p27(KIPI) or epsilon-globin gene-related histones. The HDACi caused a marked decrease in HDAC1 and Myc and an increase in RNA polymerase II in proteins bound to the p21(WAF1) promoter. Thus, this study identifies effects of SAHA on p21(WAF1)-associated proteins that explain, at least in part, the selective effect of HDACi in altering gene expression.


Assuntos
Ciclinas/genética , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Histona Desacetilases/análise , Ácidos Hidroxâmicos/farmacologia , Regiões Promotoras Genéticas , Acetilação , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21 , Desoxirribonuclease I/farmacologia , Histona Desacetilase 1 , Humanos , Processamento de Proteína Pós-Traducional , Vorinostat
5.
Cancer Res ; 61(23): 8492-7, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11731433

RESUMO

Histone deacetylase (HDACs) regulate histone acetylation by catalyzing the removal of acetyl groups on the NH(2)-terminal lysine residues of the core nucleosomal histones. Modulation of the acetylation status of core histones is involved in the regulation of the transcriptional activity of certain genes. HDAC activity is generally associated with transcriptional repression. Aberrant recruitment of HDAC activity has been associated with the development of certain human cancers. We have developed a class of HDAC inhibitors, such as suberoylanilide hydroxamic acid (SAHA), that were initially identified based on their ability to induce differentiation of cultured murine erythroleukemia cells. Additional studies have demonstrated that SAHA inhibits the growth of tumors in rodents. In this study we have examined the effects of SAHA on MCF-7 human breast cancer cells. We found that SAHA causes the inhibition of proliferation, accumulation of cells in a dose-dependent manner in G(1) then G(2)-M phase of the cell cycle, and induction of milk fat globule protein, milk fat membrane globule protein, and lipid droplets. Growth inhibition was associated with morphological changes including the flattening and enlargement of the cytoplasm, and a decrease in the nuclear:cytoplasmic ratio. Withdrawal of SAHA led to reentry of cells into the cell cycle and reversal to a less differentiated phenotype. SAHA induced differentiation in the estrogen receptor-negative cell line SKBr-3 and the retinoblastoma-negative cell line MDA-468. We propose that SAHA has profound antiproliferative activity by causing these cells to undergo cell cycle arrest and differentiation that is dependent on the presence of SAHA. SAHA and other HDAC inhibitors are currently in Phase I clinical trials. These findings may impact the clinical use of these drugs.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Inibidores Enzimáticos/farmacologia , Ácidos Hidroxâmicos/farmacologia , Neoplasias da Mama/metabolismo , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Inibidores de Histona Desacetilases , Humanos , Lipídeos/biossíntese , Proteínas do Leite/biossíntese , Receptor ErbB-2/biossíntese , Receptor ErbB-2/fisiologia , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/fisiologia , Proteína do Retinoblastoma/biossíntese , Proteína do Retinoblastoma/fisiologia , Vorinostat
6.
J Clin Invest ; 108(9): 1321-30, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11696577

RESUMO

Acute promyelocytic leukemia (APL) is associated with chromosomal translocations, invariably involving the retinoic acid receptor alpha (RAR alpha) gene fused to one of several distinct loci, including the PML or PLZF genes, involved in t(15;17) or t(11;17), respectively. Patients with t(15;17) APL respond well to retinoic acid (RA) and other treatments, whereas those with t(11;17) APL do not. The PML-RAR alpha and PLZF-RAR alpha fusion oncoproteins function as aberrant transcriptional repressors, in part by recruiting nuclear receptor-transcriptional corepressors and histone deacetylases (HDACs). Transgenic mice harboring the RAR alpha fusion genes develop forms of leukemia that faithfully recapitulate both the clinical features and the response to RA observed in humans with the corresponding translocations. Here, we investigated the effects of HDAC inhibitors (HDACIs) in vitro and in these animal models. In cells from PLZF-RAR alpha/RAR alpha-PLZF transgenic mice and cells harboring t(15;17), HDACIs induced apoptosis and dramatic growth inhibition, effects that could be potentiated by RA. HDACIs also increased RA-induced differentiation. HDACIs, but not RA, induced accumulation of acetylated histones. Using microarray analysis, we identified genes induced by RA, HDACIs, or both together. In combination with RA, all HDACIs tested overcame the transcriptional repression exerted by the RAR alpha fusion oncoproteins. In vivo, HDACIs induced accumulation of acetylated histones in target organs. Strikingly, this combination of agents induced leukemia remission and prolonged survival, without apparent toxic side effects.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/genética , Indução de Remissão , Animais , Antineoplásicos/farmacologia , Apoptose , Northern Blotting , Western Blotting , Ciclo Celular , Diferenciação Celular , Divisão Celular , DNA Complementar/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Modelos Químicos , Análise de Sequência com Séries de Oligonucleotídeos , Fenilbutiratos/farmacologia , Ligação Proteica , Receptores do Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Fatores de Tempo , Transcrição Gênica , Ativação Transcricional , Células Tumorais Cultivadas , Regulação para Cima , Vorinostat
7.
Curr Opin Oncol ; 13(6): 477-83, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11673688

RESUMO

Histone deacetylase inhibitors are potent inducers of growth arrest, differentiation, or apoptotic cell death in a variety of transformed cells in culture and in tumor bearing animals. Histone deacetylases and the family of histone acetyl transferases are involved in determining the acetylation of histones, which play a role in regulation of gene expression. Radiograph crystallographic studies reveal that the histone deacetylase inhibitors, suberoylanilide hydroxamic acid and trichostatin A, fit into the catalytic site of histone deacetylase, which has a tubular structure with a zinc atom at its base. The hydroxamic acid moiety of the inhibitor binds to the zinc. Histone deacetylase inhibitors cause acetylated histones to accumulate in both tumor and peripheral circulating mononuclear cells. Accumulation of acetylated histones has been used as a marker of the biologic activity of the agents. Hydroxamic acid-based histone deacetylase inhibitors limit tumor cell growth in animals with little or no toxicity. These compounds act selectively on genes, altering the transcription of only approximately 2% of expressed genes in cultured tumor cells. A number of proteins other than histones are substrates for histone deacetylases. The role that these other targets play in histone deacetylase inducement of cell growth arrest, differentiation, or apoptotic cell death is not known. This review summarizes the characteristics of a variety of inhibitors of histone deacetylases and their effects on transformed cells in culture and tumor growth in animal models. Several structurally different histone deacetylase inhibitors are in phase I or II clinical trials in patients with cancers.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Histonas/metabolismo , Animais , Biomarcadores Tumorais , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Neoplasias/tratamento farmacológico , Células Tumorais Cultivadas
10.
Clin Cancer Res ; 7(4): 962-70, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11309347

RESUMO

PURPOSE: We have synthesized a series of hybrid polar compounds that induce differentiation and/or apoptosis of various transformed cells. These agents are also potent inhibitors of histone deacetylases (HDACs). Pyroxamide (suberoyl-3-aminopyridineamide hydroxamic acid) is a new member of this class of compounds that is currently under development as an anticancer agent. We investigated the activity of pyroxamide as an inducer of differentiation and/or apoptosis in transformed cells. EXPERIMENTAL DESIGN AND RESULTS: Pyroxamide, at micromolar concentrations, induced terminal differentiation in murine erythroleukemia (MEL) cells and caused growth inhibition by cell cycle arrest and/or apoptosis in MEL, prostate carcinoma, bladder carcinoma, and neuroblastoma cells. Administration of pyroxamide (100 or 200 mg/kg/day) to nude mice at doses that caused little evident toxicity significantly suppressed the growth of s.c. CWR22 prostate cancer xenografts. Despite the potent growth-inhibitory effects of pyroxamide in this tumor model, serum prostate-specific antigen levels in control versus pyroxamide-treated mice were not significantly different. Pyroxamide is a potent inhibitor of affinity-purified HDAC1 (ID(50) = 100 nM) and causes the accumulation of acetylated core histones in MEL cells cultured with the agent. Human CWR22 prostate tumor xenografts from mice treated with pyroxamide (100 or 200 mg/kg/day) showed increased levels of histone acetylation and increased expression of the cell cycle regulator p21/WAF1, compared with tumors from vehicle-treated control animals. CONCLUSIONS: The findings suggest that pyroxamide may be a useful agent for the treatment of malignancy and that induction of p21/WAF1 in transformed cells by pyroxamide may contribute to the antitumor effects of this agent.


Assuntos
Aminopiridinas/farmacologia , Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/farmacologia , Acetilação/efeitos dos fármacos , Aminopiridinas/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/sangue , Neoplasias da Próstata/tratamento farmacológico , Resultado do Tratamento , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Res ; 61(9): 3591-4, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11325825

RESUMO

Histone deacetylase inhibitors (HDACIs) inhibit the growth of a variety of transformed cells in culture. We demonstrated previously that the hybrid-polar HDACI m-carboxycinnamic acid bis-hydroxamide (CBHA) induces apoptosis of human neuroblastoma in vitro and is effective in lower doses when combined with retinoids. The current study investigates the effect of CBHA on the growth of human neuroblastoma in vivo, both alone and in combination with all-trans retinoic acid (atRA), using a severe combined immunodeficiency-mouse xenograft model. CBHA (50, 100, and 200 mg/kg/day) inhibited growth of SMS-KCN-69n tumor xenografts in a dose-dependent fashion, with 200 mg/kg CBHA resulting in a complete suppression of tumor growth. The efficacy of 50 and 100 mg/kg CBHA was enhanced by the addition of 2.5 mg/kg atRA. This dose of atRA was ineffective when administered alone. Treatment was accompanied by mild weight loss in all groups except the lowest dose of CBHA. Our results suggest HDACIs alone or combined with retinoids may have therapeutic utility for neuroblastoma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Cinamatos/farmacologia , Inibidores Enzimáticos/farmacologia , Neuroblastoma/tratamento farmacológico , Tretinoína/farmacologia , Acetilação , Animais , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Divisão Celular/efeitos dos fármacos , Cinamatos/administração & dosagem , Cinamatos/toxicidade , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Inibidores Enzimáticos/toxicidade , Feminino , Inibidores do Crescimento/farmacologia , Inibidores de Histona Desacetilases , Histonas/metabolismo , Humanos , Camundongos , Camundongos SCID , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Tretinoína/administração & dosagem , Células Tumorais Cultivadas , Redução de Peso/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Nat Rev Cancer ; 1(3): 194-202, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11902574

RESUMO

Together, histone acetyltransferases and histone deacetylases (HDACs) determine the acetylation status of histones. This acetylation affects the regulation of gene expression, and inhibitors of HDACs have been found to cause growth arrest, differentiation and/or apoptosis of many tumours cells by altering the transcription of a small number of genes. HDAC inhibitors are proving to be an exciting therapeutic approach to cancer, but how do they exert this effect?


Assuntos
Histona Desacetilases/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias/enzimologia , Acetilação/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Cromatina/enzimologia , Cromatina/ultraestrutura , Ensaios Clínicos como Assunto , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/classificação , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Feminino , Previsões , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Inativação Gênica/efeitos dos fármacos , Inativação Gênica/fisiologia , Inibidores de Histona Desacetilases , Histonas/metabolismo , Humanos , Masculino , Modelos Moleculares , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/enzimologia , Nucleossomos/enzimologia , Nucleossomos/ultraestrutura , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/fisiologia , Saccharomyces cerevisiae/enzimologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Especificidade da Espécie , Fatores de Transcrição/fisiologia , Ativação Transcricional/efeitos dos fármacos
13.
Proc Natl Acad Sci U S A ; 97(26): 14329-33, 2000 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-11114188

RESUMO

Histone deacetylase 4 (HDAC4) is a member of a family of enzymes that catalyze the removal of acetyl groups from core histones, resulting in a compact chromatin structure that is generally associated with repressed gene transcription. Protein phosphorylation has been implicated in the regulation of the corepressor activity of the deacetylase. Here we report that serine/threonine kinases are found in association with HDAC4 and phosphorylate HDAC4 in vitro, and HDAC4 is phosphorylated in cells. The extracellular signal-regulated kinases 1 and 2 (ERK1/2), also known as p44(MAPK) and p42(MAPK), respectively, are two of the kinases associated with HDAC4. ERK1/2 are components of the Ras-mitogen-activated protein kinase (MAPK) signal transduction pathway. Activation of the Ras-MAPK pathway by expression of oncogenic Ras or constitutively active MAPK/ERK kinase 1 results in an increased percentage of cells (from approximately 10% to approximately 70%) that express HDAC4 in the nucleus in C2C12 myoblast cells. In cells transfected with oncogenic Ras, nuclear HDAC4 is associated with kinase activity. Our results provide evidence that protein kinase activity is present in a protein complex with HDAC4 and directly links the Ras-MAPK signal transduction pathway to a mechanism for chromatin remodeling (i.e., histone deacetylation).


Assuntos
Histona Desacetilases/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/metabolismo , Animais , Linhagem Celular , Linhagem Celular Transformada , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Ativação Enzimática , Histona Desacetilases/genética , Humanos , MAP Quinase Quinase 1 , Camundongos , Proteína Quinase 3 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Repressoras/genética
14.
Med Pediatr Oncol ; 35(6): 577-81, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11107121

RESUMO

BACKGROUND: Neuroblastoma is a common childhood cancer with a poor overall prognosis. Retinoic acids (RAs) have been studied as a potential therapy, showing promise in recurrent disease. The histone deacetylase inhibitor (HDACI) M-carboxycinnamic acid bishydroxamide (CBHA) is another potential therapy, which we recently described. Combinations of RAs and HDACIs currently under investigation display synergy in certain neoplasms. In this study, we evaluate the effect of combinations of RAs and HDACIs on human neuroblastoma cells. PROCEDURE: Established cell lines were cultured in increasing concentrations of HDACIs, RAs, and combinations thereof. Following exposure, viable cell number was quantified by trypan blue dye exclusion on a hemacytometer. Cell cycle analysis was performed by propidium iodide staining and FACS. RESULTS: All assayed HDACIs and RAs decreased viable cell number. Lower concentrations of each agent were effective when the two were combined. The primary reason for decreased cell number appears to be apoptosis following HDACI exposure and G1 arrest following RA exposure. Both effects are seen with cotreatment. Caspase inhibition abrogates the apoptotic response. CONCLUSIONS: CBHA causes apoptosis of human neuroblastoma in vitro, an effect that can add to the effects of RA. HDACIs and RAs inhibit neuroblastoma in significantly lower concentrations when used together than when used individually. Combination therapy may improve the ultimate efficacy while reducing the side effects of these agents in clinical use.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cinamatos/uso terapêutico , Inibidores de Histona Desacetilases , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Tretinoína/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Inibidores de Caspase , Divisão Celular/efeitos dos fármacos , Cinamatos/farmacologia , Fase G1/efeitos dos fármacos , Humanos , Tretinoína/farmacologia , Células Tumorais Cultivadas
15.
Cancer Res ; 60(18): 5165-70, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11016644

RESUMO

Suberoylanilide hydroxamic acid (SAHA) is the prototype of a family of hybrid polar compounds that induce growth arrest in transformed cells and show promise for the treatment of cancer. SAHA induces differentiation and/or apoptosis in certain transformed cells in culture and is a potent inhibitor of histone deacetylases. In this study, we examined the effects of SAHA on the growth of human prostate cancer cells in culture and on the growth of the CWR22 human prostate xenograft in nude mice. SAHA suppressed the growth of the LNCaP, PC-3, and TSU-Pr1 cell lines at micromolar concentrations (2.5-7.5 microM). SAHA induced dose-dependent cell death in the LNCaP cells. In mice with transplanted CWR222 human prostate tumors, SAHA (25, 50, and 100 mg/kg/day) caused significant suppression of tumor growth compared with mice receiving vehicle alone; treatment with 50 mg/kg/day resulted in a 97% reduction in the mean final tumor volume compared with controls. At this dose, there was no detectable toxicity as evaluated by weight gain and necropsy examination. Increased accumulation of acetylated core histones was detected in the CWR22 tumors within 6 h of SAHA administration. SAHA induced prostate-specific antigen mRNA expression in CWR22 prostate cancer cells, resulting in higher levels of serum prostate-specific antigen than predicted from tumor volume alone. The results suggest that hydroxamic acid-based hybrid polar compounds inhibit prostate cancer cell growth and may be useful, relatively nontoxic agents for the treatment of prostate carcinoma.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Ácidos Hidroxâmicos/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Animais , Antineoplásicos/toxicidade , Morte Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Inibidores Enzimáticos/toxicidade , Inibidores do Crescimento/farmacologia , Inibidores de Histona Desacetilases , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Transplante Heterólogo , Células Tumorais Cultivadas/efeitos dos fármacos , Vorinostat
16.
Proc Natl Acad Sci U S A ; 97(18): 10014-9, 2000 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-10954755

RESUMO

Histone deacetylases (HDACs) catalyze the removal of acetyl groups on the amino-terminal lysine residues of core nucleosomal histones. This activity is associated generally with transcriptional repression. We have reported previously that inhibition of HDAC activity by hydroxamic acid-based hybrid polar compounds, such as suberoylanilide hydroxamic acid (SAHA), induces differentiation and/or apoptosis of transformed cells in vitro and inhibits tumor growth in vivo. SAHA is a potentially new therapeutic approach to cancer treatment and is in Phase I clinical trials. In several tumor cell lines examined, HDAC inhibitors alter the expression of less than 1% of expressed genes, including the cell cycle kinase inhibitor p21(WAF1). In T24 bladder carcinoma cells, SAHA induces up to a 9-fold increase in p21(WAF1) mRNA and protein, which is, at least in part, because of an increase in the rate of transcription of the gene. SAHA causes an accumulation of acetylated histones H3 and H4 in total cellular chromatin by 2 h, which is maintained through 24 h of culture. An increase in the accumulation of acetylated H3 and H4 was detected throughout the p21(WAF1) promoter and the structural gene after culture with SAHA. The level of histone acetylation did not change in chromatin associated with the actin and p27 genes, and their mRNA expression was not altered during culture of T24 cells with SAHA. Thus, the present findings indicate that the induction of p21(WAF1) by SAHA is regulated, at least in part, by the degree of acetylation of the gene-associated histones and that this induced increase in acetylation is gene selective.


Assuntos
Anticarcinógenos/farmacologia , Ciclinas/genética , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Ácidos Hidroxâmicos/farmacologia , Transcrição Gênica , Núcleo Celular/metabolismo , Cromatina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Primers do DNA , Inibidores Enzimáticos/metabolismo , Regulação Enzimológica da Expressão Gênica , Inibidores de Histona Desacetilases , Histonas/metabolismo , Humanos , Reação em Cadeia da Polimerase , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária , Vorinostat
17.
J Natl Cancer Inst ; 92(15): 1210-6, 2000 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-10922406

RESUMO

Histone deacetylase (HDAC) inhibitors have been shown to be potent inducers of growth arrest, differentiation, and/or apoptotic cell death of transformed cells in vitro and in vivo. One class of HDAC inhibitors, hydroxamic acid-based hybrid polar compounds (HPCs), induce differentiation at micromolar or lower concentrations. Studies (x-ray crystallographic) showed that the catalytic site of HDAC has a tubular structure with a zinc atom at its base and that these HDAC inhibitors, such as suberoylanilide hydroxamic acid and trichostatin A, fit into this structure with the hydroxamic moiety of the inhibitor binding to the zinc. HDAC inhibitors cause acetylated histones to accumulate in both tumor and normal tissues, and this accumulation can be used as a marker of the biologic activity of the HDAC inhibitors. Hydroxamic acid-based HPCs act selectively to inhibit tumor cell growth at levels that have little or no toxicity for normal cells. These compounds also act selectively on gene expression, altering the expression of only about 2% of the genes expressed in cultured tumor cells. In general, chromatin fractions enriched in actively transcribed genes are also enriched in highly acetylated core histones, whereas silent genes are associated with nucleosomes with a low level of acetylation. However, HDACs can also acetylate proteins other than histones in nucleosomes. The role that these other targets play in the induction of cell growth arrest, differentiation, and/or apoptotic cell death has not been determined. Our working hypothesis is that inhibition of HDAC activity leads to the modulation of expression of a specific set of genes that, in turn, result in growth arrest, differentiation, and/or apoptotic cell death. The hydroxamic acid-based HPCs are potentially effective agents for cancer therapy and, possibly, cancer chemoprevention.


Assuntos
Acetiltransferases/metabolismo , Apoptose/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Neoplasias/tratamento farmacológico , Proteínas de Saccharomyces cerevisiae , Animais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histona Acetiltransferases , Humanos , Neoplasias/enzimologia
18.
Oncogene ; 18(50): 7016-25, 1999 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-10597302

RESUMO

Determinants of differentiation and apoptosis in myelomonocytic leukemia cells (U937) exposed to the novel hybrid polar compound SAHA (suberoylanilide hydroxamic acid) have been examined. In contrast to hexamethylenbisacetamide (HMBA), SAHA-related maturation was limited and accompanied by marked cytoxicity. SAHA-mediated apoptosis occurred within the G0G1 and S phase populations, and was associated with decreased mitochondrial membrane potential, caspase-3 activation, PARP degradation, hypophosphorylation/cleavage of pRB, and down-regulation of c-Myc, c-Myb, and B-Myb. Enforced expression of Bcl-2 or Bcl-XL inhibited SAHA-induced apoptosis, but only modestly potentiated differentiation. While SAHA induced the cyclin-dependent kinase inhibitor p21CIP1, antisense ablation of this CDKI increased, rather than decreased, SAHA-related lethality. In contrast, conditional expression of wild-type p53 failed to modify SAHA actions, but markedly potentiated HMBA-induced apoptosis. Finally, SAHA modestly increased expression/activation of the stress-activated protein kinase (SAPK/JNK); moreover, SAHA-related lethality was partially attenuated by a dominant-negative c-Jun mutant protein (TAM67). SAHA did not stimulate mitogen-activated protein kinase (MAPK), nor was lethality diminished by the specific MEK/MAPK inhibitor PD98059. These findings indicate that SAHA potently induces apoptosis in human leukemia cells via a pathway that is p53-independent but at least partially regulated by Bcl-2/Bcl-XL, p21CIP1, and the c-Jun/AP-1 signaling cascade.


Assuntos
Apoptose/efeitos dos fármacos , Ciclinas/metabolismo , Ácidos Hidroxâmicos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Regulação para Baixo , Humanos , Leucemia/metabolismo , Leucemia/patologia , Proteínas Proto-Oncogênicas c-myb/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Células U937 , Vorinostat , Proteína bcl-X
19.
Nature ; 401(6749): 188-93, 1999 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-10490031

RESUMO

Histone deacetylases (HDACs) mediate changes in nucleosome conformation and are important in the regulation of gene expression. HDACs are involved in cell-cycle progression and differentiation, and their deregulation is associated with several cancers. HDAC inhibitors, such as trichostatin A (TSA) and suberoylanilide hydroxamic acid (SAHA), have anti-tumour effects, as they can inhibit cell growth, induce terminal differentiation and prevent the formation of tumours in mice models, and they are effective in the treatment of promyelocytic leukemia. Here we describe the structure of the histone deacetylase catalytic core, as revealed by the crystal structure of a homologue from the hyperthermophilic bacterium Aquifex aeolicus, that shares 35.2% identity with human HDAC1 over 375 residues, deacetylates histones in vitro and is inhibited by TSA and SAHA. The deacetylase, deacetylase-TSA and deacetylase-SAHA structures reveal an active site consisting of a tubular pocket, a zinc-binding site and two Asp-His charge-relay systems, and establish the mechanism of HDAC inhibition. The residues that make up the active site and contact the inhibitors are conserved across the HDAC family. These structures also suggest a mechanism for the deacetylation reaction and provide a framework for the further development of HDAC inhibitors as antitumour agents.


Assuntos
Inibidores Enzimáticos/química , Histona Desacetilases/química , Ácidos Hidroxâmicos/química , Sequência de Aminoácidos , Domínio Catalítico , Cristalografia por Raios X , Inibidores Enzimáticos/metabolismo , Escherichia coli , Bacilos e Cocos Aeróbios Gram-Negativos/química , Bacilos e Cocos Aeróbios Gram-Negativos/enzimologia , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes de Fusão/química , Homologia de Sequência de Aminoácidos , Vorinostat , Zinco/química
20.
Cancer Res ; 59(17): 4392-9, 1999 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10485488

RESUMO

Inhibitors of histone deacetylase (HDAC) have been shown to have both apoptotic and differentiating effects on various tumor cells. M-carboxycinnamic acid bishydroxamide (CBHA) is a recently developed hybrid polar compound structurally related to hexamethylene bisacetamide. CBHA is a potent inhibitor of HDAC activity. CBHA induces cellular growth arrest and differentiation in model tumor systems. We undertook an investigation of the effects of CBHA on human neuroblastoma cell lines in vitro. When added to cultures of a panel of neuroblastoma cell lines, CBHA induced the accumulation of acetylated histones H3 and H4, consistent with the inhibition of HDAC. Concentrations of CBHA between 0.5 microM and 4 microM led to apoptosis in nine of nine neuroblastoma cell lines. Apoptosis was assessed by DNA fragmentation analysis and the appearance of a sub-G1 (<2N ploidy) population by flow cytometric analysis. The addition of a caspase inhibitor (benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone) completely abrogated CBHA-induced apoptosis in three of three cell lines. The addition of cycloheximide greatly reduced CBHA-induced apoptosis, suggesting that apoptotic induction was dependent on de novo protein synthesis. In addition, CBHA induced the expression of both CD95 (APO-1/Fas) and CD95 ligand within 12 h. The effect of CBHA on human neuroblastoma cells suggests that this agent and structurally related synthetic hybrid polar compounds have therapeutic potential for the treatment of this malignancy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Cinamatos/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Glicoproteínas de Membrana/biossíntese , Neuroblastoma/tratamento farmacológico , Receptor fas/biossíntese , Inibidores de Caspase , Divisão Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Cicloeximida/farmacologia , Fragmentação do DNA/efeitos dos fármacos , Proteína Ligante Fas , Histonas/metabolismo , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA