Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 14: 1135181, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37214253

RESUMO

Osteoporosis is an age-related disease of bone metabolism marked by reduced bone mineral density and impaired bone strength. The disease causes the bones to weaken and break more easily. Osteoclasts participate in bone resorption more than osteoblasts participate in bone formation, disrupting bone homeostasis and leading to osteoporosis. Currently, drug therapy for osteoporosis includes calcium supplements, vitamin D, parathyroid hormone, estrogen, calcitonin, bisphosphates, and other medications. These medications are effective in treating osteoporosis but have side effects. Copper is a necessary trace element in the human body, and studies have shown that it links to the development of osteoporosis. Cuproptosis is a recently proposed new type of cell death. Copper-induced cell death regulates by lipoylated components mediated via mitochondrial ferredoxin 1; that is, copper binds directly to the lipoylated components of the tricarboxylic acid cycle, resulting in lipoylated protein accumulation and subsequent loss of iron-sulfur cluster proteins, leading to proteotoxic stress and eventually cell death. Therapeutic options for tumor disorders include targeting the intracellular toxicity of copper and cuproptosis. The hypoxic environment in bone and the metabolic pathway of glycolysis to provide energy in cells can inhibit cuproptosis, which may promote the survival and proliferation of various cells, including osteoblasts, osteoclasts, effector T cells, and macrophages, thereby mediating the osteoporosis process. As a result, our group tried to explain the relationship between the role of cuproptosis and its essential regulatory genes, as well as the pathological mechanism of osteoporosis and its effects on various cells. This study intends to investigate a new treatment approach for the clinical treatment of osteoporosis that is beneficial to the treatment of osteoporosis.


Assuntos
Apoptose , Reabsorção Óssea , Osteoporose , Humanos , Reabsorção Óssea/metabolismo , Cobre , Osteoclastos/metabolismo , Osteoporose/patologia , Hormônio Paratireóideo/metabolismo
2.
Stem Cell Rev Rep ; 17(6): 2304-2313, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34561772

RESUMO

Polycomb group protein Bmi1 is essential for hematopoietic stem cell (HSC) self-renewal and terminal differentiation. However, its target genes in hematopoietic stem and progenitor cells are largely unknown. We performed gene expression profiling assays and found that genes of the Wnt signaling pathway are significantly elevated in Bmi1 null hematopoietic stem and progenitor cells (HSPCs). Bmi1 is associated with several genes of the Wnt signaling pathway in hematopoietic cells. Further, we found that Bmi1 represses Wnt gene expression in HSPCs. Importantly, loss of ß-catenin, which reduces Wnt activation, partially rescues the HSC self-renewal and differentiation defects seen in the Bmi1 null mice. Thus, we have identified Bmi1 as a novel regulator of Wnt signaling pathway in HSPCs. Given that Wnt signaling pathway plays an important role in hematopoiesis, our studies suggest that modulating Wnt signaling may hold potential for enhancing HSC self-renewal, thereby improving the outcomes of HSC transplantation.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Via de Sinalização Wnt , Animais , Hematopoese/genética , Células-Tronco Hematopoéticas , Camundongos , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Via de Sinalização Wnt/genética
3.
Int J Mol Sci ; 21(22)2020 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-33202645

RESUMO

Polycomb group (PcG) proteins function as vital epigenetic regulators in various biological processes, including pluripotency, development, and carcinogenesis. PcG proteins form multicomponent complexes, and two major types of protein complexes have been identified in mammals to date, Polycomb Repressive Complexes 1 and 2 (PRC1 and PRC2). The PRC1 complexes are composed in a hierarchical manner in which the catalytic core, RING1A/B, exclusively interacts with one of six Polycomb group RING finger (PCGF) proteins. This association with specific PCGF proteins allows for PRC1 to be subdivided into six distinct groups, each with their own unique modes of action arising from the distinct set of associated proteins. Historically, PRC1 was considered to be a transcription repressor that deposited monoubiquitylation of histone H2A at lysine 119 (H2AK119ub1) and compacted local chromatin. More recently, there is increasing evidence that demonstrates the transcription activation role of PRC1. Moreover, studies on the higher-order chromatin structure have revealed a new function for PRC1 in mediating long-range interactions. This provides a different perspective regarding both the transcription activation and repression characteristics of PRC1. This review summarizes new advancements regarding the composition of mammalian PRC1 and accompanying explanations of how diverse PRC1-associated proteins participate in distinct transcription regulation mechanisms.


Assuntos
Cromatina/metabolismo , Histonas/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Transcrição Gênica/fisiologia , Ubiquitinação/fisiologia , Animais , Cromatina/genética , Histonas/genética , Humanos , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 2/genética
4.
Nat Commun ; 10(1): 5649, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31827082

RESUMO

Clonal hematopoiesis of indeterminate potential (CHIP) increases with age and is associated with increased risks of hematological malignancies. While TP53 mutations have been identified in CHIP, the molecular mechanisms by which mutant p53 promotes hematopoietic stem and progenitor cell (HSPC) expansion are largely unknown. Here we discover that mutant p53 confers a competitive advantage to HSPCs following transplantation and promotes HSPC expansion after radiation-induced stress. Mechanistically, mutant p53 interacts with EZH2 and enhances its association with the chromatin, thereby increasing the levels of H3K27me3 in genes regulating HSPC self-renewal and differentiation. Furthermore, genetic and pharmacological inhibition of EZH2 decreases the repopulating potential of p53 mutant HSPCs. Thus, we uncover an epigenetic mechanism by which mutant p53 drives clonal hematopoiesis. Our work will likely establish epigenetic regulator EZH2 as a novel therapeutic target for preventing CHIP progression and treating hematological malignancies with TP53 mutations.


Assuntos
Epigênese Genética , Doenças Hematológicas/metabolismo , Hematopoese , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Feminino , Doenças Hematológicas/genética , Doenças Hematológicas/fisiopatologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Metilação , Camundongos Endogâmicos C57BL , Mutação , Ligação Proteica
5.
J Org Chem ; 83(24): 15225-15235, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30468074

RESUMO

Herein, we report an enantioselective synthesis of azepinones via the N-heterocyclic carbene (NHC) catalyzed [3+4] annulation reaction of isatin-derived enals and aurone-derived azadienes. The corresponding spirocyclic oxindole-benzofuroazepinones were obtained in good yields, with excellent diastereo- and enantioselectivities. The resulted azepinones were evaluated for their in vitro cytotoxic activities against six human tumor cell lines, with two compounds showing significant inhibitory activity comparable with that of cisplatin.

6.
Nat Prod Commun ; 11(5): 667-70, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27319146

RESUMO

Four new compounds, a dibenzylbutane lignin, plasiaticine F (1), an acetylenic glycoside, plasiaticine G (2), an indole alkaloid, plasiaticine H (3), and an ionone derivative, plasiaticine I (4), were isolated from the seeds of Plantago asiatica. The structures of the new compounds were elucidated on the basis of comprehensive analysis of spectroscopic data. Compounds 1-3 were tested for their cytotoxicity, but lacked significant activity.


Assuntos
Lignina/análogos & derivados , Plantago/química , Ensaios de Seleção de Medicamentos Antitumorais , Células HL-60 , Humanos , Lignina/química , Lignina/isolamento & purificação , Células MCF-7 , Estrutura Molecular , Sementes/química
7.
Sci Rep ; 6: 24945, 2016 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-27101899

RESUMO

Plasiatine (1), isolated from the seeds of Plantago asiatica, is an unprecedented indole analogue linked to a phenylpropanoid moiety via a carbon bond that builds up a novel heteromeric construction with a C19N2 scaffold. Its structure was determined by spectroscopic data and computational evidence. Notably, experimental assay demonstrated that 1 significantly enhanced the activity of the nonreceptor protein tyrosine phosphatase Shp2 in vitro in a concentration-dependent manner with an EC50 value of 0.97 µM, and activated phosphorylation of ERK, a known target of Shp2. Moreover, plasiatine (1) promoted hepatocellular HepG2 cells migration. Molecular docking suggested that plasiatine (1) binds to the catalytic cleft of Shp2. These results identified plasiatine (1) as the first small molecule Shp2 activator, and it warrants further investigation as a novel pharmaceutical tool to study the function of Shp2 in tumorigenesis.


Assuntos
Produtos Biológicos/farmacologia , Ativadores de Enzimas/farmacologia , Indóis/farmacologia , Extratos Vegetais/farmacologia , Plantago/química , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Produtos Biológicos/isolamento & purificação , Movimento Celular/efeitos dos fármacos , Ativadores de Enzimas/isolamento & purificação , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/fisiologia , Humanos , Indóis/isolamento & purificação , Simulação de Acoplamento Molecular , Estrutura Molecular , Fosforilação , Extratos Vegetais/isolamento & purificação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Sementes/química , Análise Espectral
8.
Arch Pharm Res ; 37(2): 168-74, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23703254

RESUMO

Two new triterpenoids, schisphendilactone A and B (1 and 2), together with three known triterpenoids, were isolated from the stems of Schisandra sphenanthera. Their structures were elucidated by spectroscopic methods, and the absolute configuration of 1 was determined by single-crystal X-ray diffraction. Compound 2 showed moderate inhibitory activity against SW480 cancer cell line, and compound 5 exhibited promising anti-HIV-1 activity with EC50 value of 0.52 µg ml(-1) and therapeutic index value of 117.12.


Assuntos
Fármacos Anti-HIV , Antineoplásicos Fitogênicos , Medicamentos de Ervas Chinesas , Schisandra/química , Triterpenos , Fármacos Anti-HIV/isolamento & purificação , Fármacos Anti-HIV/farmacologia , Antineoplásicos Fitogênicos/isolamento & purificação , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Efeito Citopatogênico Viral , Medicamentos de Ervas Chinesas/isolamento & purificação , Medicamentos de Ervas Chinesas/farmacologia , Humanos , Concentração Inibidora 50 , Estrutura Molecular , Caules de Planta/química , Estereoisomerismo , Triterpenos/isolamento & purificação , Triterpenos/farmacologia
9.
Mol Cell ; 45(3): 344-56, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22325352

RESUMO

The heterogeneous nature of mammalian PRC1 complexes has hindered our understanding of their biological functions. Here, we present a comprehensive proteomic and genomic analysis that uncovered six major groups of PRC1 complexes, each containing a distinct PCGF subunit, a RING1A/B ubiquitin ligase, and a unique set of associated polypeptides. These PRC1 complexes differ in their genomic localization, and only a small subset colocalize with H3K27me3. Further biochemical dissection revealed that the six PCGF-RING1A/B combinations form multiple complexes through association with RYBP or its homolog YAF2, which prevents the incorporation of other canonical PRC1 subunits, such as CBX, PHC, and SCM. Although both RYBP/YAF2- and CBX/PHC/SCM-containing complexes compact chromatin, only RYBP stimulates the activity of RING1B toward H2AK119ub1, suggesting a central role in PRC1 function. Knockdown of RYBP in embryonic stem cells compromised their ability to form embryoid bodies, likely because of defects in cell proliferation and maintenance of H2AK119ub1 levels.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Repressoras/fisiologia , Diferenciação Celular , Proliferação de Células , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Corpos Embrioides/metabolismo , Expressão Gênica , Células HEK293 , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Complexos Multiproteicos/genética , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Nucleossomos/metabolismo , Nucleossomos/ultraestrutura , Complexo Repressor Polycomb 1 , Proteínas do Grupo Polycomb , Regiões Promotoras Genéticas , Ligação Proteica , Proteômica , Proteínas Repressoras/genética , Homologia de Sequência de Aminoácidos , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
10.
J Med Chem ; 53(24): 8734-46, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-21080724

RESUMO

We have shown previously that the target of the potent cytotoxic agent 4-[(7-bromo-2-methyl-4-oxo-3H-quinazolin-6-yl)methyl-prop-2-ynylamino]-N-(3-pyridylmethyl)benzamide (CB38065, 1) is nicotinamide phosphoribosyltransferase (Nampt). With its cellular target known we sought to optimize the biochemical and cellular Nampt activity of 1 as well as its cytotoxicity. It was found that a 3-pyridylmethylamide substituent in the A region was critical to cellular Nampt activity and cytotoxicity, although other aromatic substitution did yield compounds with submicromolar enzymatic inhibition. Small unsaturated groups worked best in the D-region of the molecule, with 3,3-dimethylallyl providing optimal potency. The E region required a quinazolin-4-one or 1,2,3-benzotriazin-4-one group for activity, and many substituents were tolerated at C² of the quinazolin-4-one. The best compounds showed subnanomolar inhibition of Nampt and low nanomolar cytotoxicity in cellular assays.


Assuntos
Antineoplásicos/síntese química , Benzamidas/síntese química , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Quinazolinas/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Benzamidas/química , Benzamidas/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Células HCT116 , Humanos , Modelos Moleculares , Quinazolinas/química , Quinazolinas/farmacologia , Relação Estrutura-Atividade
11.
Chem Biol ; 17(6): 659-64, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20609415

RESUMO

Drug discovery based on cellular phenotypes is impeded by the challenge of identifying the molecular target. To alleviate this problem, we developed a chemical proteomic process to identify cellular proteins that bind to small molecules. CB30865 is a potent (subnanomolar) and selective cytotoxic compound of previously unknown mechanism of action. By combining chemical proteomics with biochemical and cellular pharmacology we have determined that CB30865 cytotoxicity is due to subnanomolar inhibition of nicotinamide phosphoribosyltransferase (Nampt), an enzyme present in the NAD biosynthetic pathway. Cancer cells develop dependence on Nampt due to increased energy requirements and the elevated activity of NAD consuming enzymes such as sirtuins and mono and poly(ADP-ribose) polymerases (PARPs). These findings suggest new chemical starting points for Nampt inhibitors and further implicate this enzyme as a target in cancer.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Nicotinamida Fosforribosiltransferase/metabolismo , Produção de Droga sem Interesse Comercial , Proteômica/métodos , Quinazolinas/metabolismo , Quinazolinas/farmacologia , Antineoplásicos/química , Descoberta de Drogas , Células HCT116 , Humanos , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Quinazolinas/química
12.
Bioorg Med Chem Lett ; 20(5): 1677-9, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20137940

RESUMO

Valosin-containing protein (VCP; also known as p97) is a member of the AAA ATPase family with a central role in the ubiquitin-degradation of misfolded proteins. VCP also exhibits antiapoptotic function and metastasis via activation of nuclear factor kappa-B signaling pathway. We have discovered that 2-anilino-4-aryl-1,3-thiazoles are potent drug-like inhibitors of this enzyme. The identified compounds show low nanomolar VCP potency, demonstrate SAR trends, and show activity in a mechanism based cellular assay. This series of compounds represents the first steps towards a novel, small molecule VCP inhibitor as a cancer therapeutic.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Compostos de Anilina/química , Antineoplásicos/química , Proteínas de Ciclo Celular/antagonistas & inibidores , Tiazóis/química , Adenosina Trifosfatases/metabolismo , Compostos de Anilina/síntese química , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Células HeLa , Humanos , NF-kappa B/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade , Tiazóis/síntese química , Tiazóis/farmacologia , Proteína com Valosina
13.
Autophagy ; 6(1): 126-37, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20061800

RESUMO

Ubiquitin-proteasome system and autophagy are the two major mechanisms for protein degradation in eukaryotic cells. LC3, a ubiquitin-like protein, plays an essential role in autophagy through its ability to be conjugated to phosphatidylethanolamine. In this study, we discovered a novel LC3-processing activity, and biochemically purified the 20S proteasome as the responsible enzyme. Processing of LC3 by the 20S proteasome is ATP- and ubiquitin-independent, and requires both the N-terminal helices and the ubiquitin fold of LC3; addition of the N-terminal helices of LC3 to the N terminus of ubiquitin renders ubiquitin susceptible to 20S proteasomal activity. Further, the 20S proteasome processes LC3 in a stepwise manner, it first cleaves LC3 within its ubiquitin fold and thus disrupts the conjugation function of LC3; subsequently and especially at high concentrations of the proteasome, LC3 is completely degraded. Intriguingly, proteolysis of LC3 by the 20S proteasome can be inhibited by p62, an LC3-binding protein that mediates autophagic degradation of polyubiquitin aggregates in cells. Therefore, our study implicates a potential mechanism underlying interplay between the proteasomal and autophagic pathways. This study also provides biochemical evidence suggesting relevance of the controversial ubiquitin-independent proteolytic activity of the 20S proteasome.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Extratos Celulares/farmacologia , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Células HeLa , Humanos , Leupeptinas/farmacologia , Camundongos , Proteínas Associadas aos Microtúbulos/química , Complexo de Endopeptidases do Proteassoma/fisiologia , Inibidores de Proteassoma , Ligação Proteica , Dobramento de Proteína/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estrutura Terciária de Proteína/efeitos dos fármacos , Proteína Sequestossoma-1 , Especificidade por Substrato , Ubiquitina/química , Ubiquitina/metabolismo , Ubiquitinação/efeitos dos fármacos
14.
J Biol Chem ; 282(42): 30718-27, 2007 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17724022

RESUMO

Caspase activation, the executing event of apoptosis, is under deliberate regulation. IAP proteins inhibit caspase activity, whereas Smac/Diablo antagonizes IAP. XIAP, a ubiquitous IAP, can inhibit both caspase-9, the initiator caspase of the mitochondrial apoptotic pathway, and the downstream effector caspases, caspase-3 and caspase-7. Smac neutralizes XIAP inhibition of caspase-9 by competing for binding of the BIR3 domain of XIAP with caspase-9, whereas how Smac liberates effector caspases from XIAP inhibition is not clear. It is generally believed that binding of Smac with IAP generates a steric hindrance that prevents XIAP from inhibiting effector caspases, and therefore small molecule mimics of Smac are not able to reverse inhibition of the effector caspases. Surprisingly, we show here that binding of a dimeric Smac N-terminal peptide with the BIR2 domain of XIAP effectively antagonizes inhibition of caspase-3 by XIAP. Further, we defined the dynamic and cooperative interaction of Smac with XIAP: binding of Smac with the BIR3 domain anchors the subsequent binding of Smac with the BIR2 domain, which in turn attenuates the caspase-3 inhibitory function of XIAP. We also show that XIAP homotrimerizes via its C-terminal Ring domain, making its inhibitory activity toward caspase-3 more susceptible to Smac.


Assuntos
Apoptose/fisiologia , Inibidores de Caspase , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Mitocondriais/metabolismo , Peptídeos/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Proteínas Reguladoras de Apoptose , Caspase 3/química , Caspase 3/metabolismo , Caspase 7/química , Caspase 7/metabolismo , Caspase 9/química , Caspase 9/metabolismo , Dimerização , Ativação Enzimática/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Mitocôndrias/química , Mitocôndrias/enzimologia , Proteínas Mitocondriais/química , Peptídeos/química , Estrutura Terciária de Proteína/fisiologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/química
15.
Cell ; 128(1): 129-39, 2007 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-17218260

RESUMO

The tumor suppressor PTEN, a critical regulator for multiple cellular processes, is mutated or deleted frequently in various human cancers. Subtle reductions in PTEN expression levels have profound impacts on carcinogenesis. Here we show that PTEN level is regulated by ubiquitin-mediated proteasomal degradation, and purified its ubiquitin ligase as HECT-domain protein NEDD4-1. In cells NEDD4-1 negatively regulates PTEN stability by catalyzing PTEN polyubiquitination. Consistent with the tumor-suppressive role of PTEN, overexpression of NEDD4-1 potentiated cellular transformation. Strikingly, in a mouse cancer model and multiple human cancer samples where the genetic background of PTEN was normal but its protein levels were low, NEDD4-1 was highly expressed, suggesting that aberrant upregulation of NEDD4-1 can posttranslationally suppress PTEN in cancers. Elimination of NEDD4-1 expression inhibited xenotransplanted tumor growth in a PTEN-dependent manner. Therefore, NEDD4-1 is a potential proto-oncogene that negatively regulates PTEN via ubiquitination, a paradigm analogous to that of Mdm2 and p53.


Assuntos
PTEN Fosfo-Hidrolase/metabolismo , Proto-Oncogenes , Ubiquitina-Proteína Ligases/metabolismo , Animais , Transformação Celular Neoplásica , Complexos Endossomais de Distribuição Requeridos para Transporte , Células HeLa , Humanos , Camundongos , Ubiquitina-Proteína Ligases Nedd4 , Transplante de Neoplasias , Neoplasias/patologia , Poliubiquitina/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proto-Oncogene Mas , Interferência de RNA , Especificidade por Substrato , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/isolamento & purificação
16.
J Biol Chem ; 280(46): 38271-5, 2005 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-16172118

RESUMO

Caspase-2 is an initiating caspase required for stress-induced apoptosis in various human cancer cells. Recent studies suggest that it can mediate the death function of tumor suppressor p53 and is activated by a multimeric protein complex, PIDDosome. However, it is not clear how caspase-2 exerts its apoptotic function in cells and whether its enzymatic activity is required for the apoptotic function. In this study, we used both in vitro mitochondrial cytochrome c release assays and cell culture apoptosis analyses to investigate the mechanism by which caspase-2 induces apoptosis. We show that active caspase-2, but neither a catalytically mutated caspase-2 nor active caspase-2 with its inhibitor, can cause cytochrome c release. Caspase-2 failed to induce cytochrome c release from mitochondria with Bid(-/-) background, and the release could be restored by addition of the wild-type Bid protein, but not by Bid with the caspase-2 cleavage site mutated. Caspase-2 was not able to induce cytochrome c release from Bax(-/-)Bak(-/-) mitochondria either. In cultured cells, gene deletion of Bax/Bak or Bid abrogated apoptosis induced by overexpression of caspase-2. Collectively, these results indicate that proteolytic activation of Bid and the subsequent induction of the mitochondrial apoptotic pathway through Bax/Bak is essential for apoptosis triggered by caspase-2.


Assuntos
Apoptose , Caspases/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Caspase 2 , Caspases/metabolismo , Catálise , Linhagem Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Citocromos/metabolismo , Citocromos c/metabolismo , DNA Complementar/metabolismo , Ativação Enzimática , Escherichia coli/metabolismo , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mutagênese Sítio-Dirigida , Plasmídeos/metabolismo , Interferência de RNA , Proteína Supressora de Tumor p53/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
17.
Proc Natl Acad Sci U S A ; 101(52): 18030-5, 2004 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-15596714

RESUMO

Histone deacetylase (HDAC) inhibitors can induce programmed cell death in cancer cells, although the underlying mechanism is obscure. In this study, we show that two distinct HDAC inhibitors, butyrate and suberoylanilide hydroxamic acid (SAHA), induced caspase-3 activation and cell death in multiple human cancer cell lines. The activation of caspase-3 was via the mitochondria/cytochrome c-mediated apoptotic pathway because it was abrogated in mouse embryonic fibroblasts with knockout of Apaf-1, the essential mediator of the pathway. Overexpression of Bcl-XL in HeLa cells also blocked caspase activation by the HDAC inhibitors. Nevertheless, Apaf-1 knockout, overexpression of Bcl-XL, and pharmacological inhibition of caspase activity did not prevent SAHA and butyrate-induced cell death. The cells undergoing such caspase-independent death had unambiguous morphological features of autophagic cell death. Therefore, HDAC inhibitors can induce both mitochondria-mediated apoptosis and caspase-independent autophagic cell death. Induction of autophagic cell death by HDAC inhibitors has clear clinical implications in treating cancers with apoptotic defects.


Assuntos
Apoptose , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Animais , Fator Apoptótico 1 Ativador de Proteases , Autofagia , Butiratos/farmacologia , Caspase 3 , Caspases/metabolismo , Morte Celular , Linhagem Celular Tumoral , Citocromos c/metabolismo , Citosol/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Fibroblastos/metabolismo , Células HeLa , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Mitocôndrias/patologia , Neoplasias/patologia , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Frações Subcelulares , Fatores de Tempo , Vorinostat , Proteína bcl-X
18.
Proc Natl Acad Sci U S A ; 99(3): 1182-7, 2002 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-11818547

RESUMO

Wnt signaling plays a key role in cell proliferation and development. Recently, casein kinase I (CKI) and protein phosphatase 2A (PP2A) have emerged as positive and negative regulators of the Wnt pathway, respectively. However, it is not clear how these two enzymes with opposing functions regulate Wnt signaling. Here we show that both CKI delta and CKI epsilon interacted directly with Dvl-1, and that CKI phosphorylated multiple components of the Wnt-regulated beta-catenin degradation complex in vitro, including Dvl-1, adenomatous polyposis coli (APC), axin, and beta-catenin. Comparison of peptide maps from in vivo and in vitro phosphorylated beta-catenin and axin suggests that CKI phosphorylates these proteins in vivo as well. CKI abrogated beta-catenin degradation in Xenopus egg extracts. Notably, CKI decreased, whereas inhibition of CKI increased, the association of PP2A with the beta-catenin degradation complex in vitro. Additionally, inhibition of CKI in vivo stabilized the beta-catenin degradation complex, suggesting that CKI actively destabilizes the complex in vivo. The ability of CKI to induce secondary body axes in Xenopus embryos was reduced by the B56 regulatory subunit of PP2A, and kinase-dead CKI epsilon acted synergistically with B56 in inhibiting Wnt signaling. The data suggest that CKI phosphorylates and destabilizes the beta-catenin degradation complex, likely through the dissociation of PP2A, providing a mechanism by which CKI stabilizes beta-catenin and propagates the Wnt signal.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas Quinases/metabolismo , Proteínas Repressoras , Transativadores , Proteínas de Peixe-Zebra , Animais , Proteína Axina , Caderinas/química , Caderinas/metabolismo , Caseína Quinases , Linhagem Celular , Clonagem Molecular , Proteínas do Citoesqueleto/química , Primers do DNA , Feminino , Humanos , Cinética , Oócitos/metabolismo , Mapeamento de Peptídeos , Fosfopeptídeos/química , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Biossíntese de Proteínas , Proteínas Quinases/isolamento & purificação , Proteína Fosfatase 2 , Proteínas Tirosina Quinases/metabolismo , Proteínas/química , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Transcrição Gênica , Transfecção , Proteínas Wnt , Proteínas de Xenopus , Xenopus laevis , beta Catenina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA