Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Biol Chem ; 289(48): 33425-41, 2014 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-25305013

RESUMO

Artesunate (ART) is an anti-malaria drug that has been shown to exhibit anti-tumor activity, and functional lysosomes are reported to be required for ART-induced cancer cell death, whereas the underlying molecular mechanisms remain largely elusive. In this study, we aimed to elucidate the molecular mechanisms underlying ART-induced cell death. We first confirmed that ART induces apoptotic cell death in cancer cells. Interestingly, we found that ART preferably accumulates in the lysosomes and is able to activate lysosomal function via promotion of lysosomal V-ATPase assembly. Furthermore, we found that lysosomes function upstream of mitochondria in reactive oxygen species production. Importantly, we provided evidence showing that lysosomal iron is required for the lysosomal activation and mitochondrial reactive oxygen species production induced by ART. Finally, we showed that ART-induced cell death is mediated by the release of iron in the lysosomes, which results from the lysosomal degradation of ferritin, an iron storage protein. Meanwhile, overexpression of ferritin heavy chain significantly protected cells from ART-induced cell death. In addition, knockdown of nuclear receptor coactivator 4, the adaptor protein for ferritin degradation, was able to block ART-mediated ferritin degradation and rescue the ART-induced cell death. In summary, our study demonstrates that ART treatment activates lysosomal function and then promotes ferritin degradation, subsequently leading to the increase of lysosomal iron that is utilized by ART for its cytotoxic effect on cancer cells. Thus, our data reveal a new mechanistic action underlying ART-induced cell death in cancer cells.


Assuntos
Antimaláricos/farmacologia , Artemisininas/farmacologia , Ferritinas/metabolismo , Lisossomos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Proteólise/efeitos dos fármacos , Artesunato , Morte Celular/efeitos dos fármacos , Células HeLa , Células Hep G2 , Humanos , Ferro/metabolismo , Neoplasias/metabolismo , Neoplasias/fisiopatologia , Coativadores de Receptor Nuclear/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
2.
Angew Chem Int Ed Engl ; 54(8): 2515-9, 2015 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-25565365

RESUMO

Poly(ADP-ribose)polymerase-1 (PARP1) is a BRCT-containing enzyme (BRCT = BRCA1 C-terminus) mainly involved in DNA repair and damage response and a validated target for cancer treatment. Small-molecule inhibitors that target the PARP1 catalytic domain have been actively pursued as anticancer drugs, but are potentially problematic owing to a lack of selectivity. Compounds that are capable of disrupting protein-protein interactions of PARP1 provide an alternative by inhibiting its activities with improved selectivity profiles. Herein, by establishing a high-throughput microplate-based assay suitable for screening potential PPI inhibitors of the PARP1 BRCT domain, we have discovered that (±)-gossypol, a natural product with a number of known biological activities, possesses novel PARP1 inhibitory activity both in vitro and in cancer cells and presumably acts through disruption of protein-protein interactions. As the first known cell-permeable small-molecule PPI inhibitor of PAPR1, we further established that (-)-gossypol was likely the causative agent of PARP1 inhibition by promoting the formation of a 1:2 compound/PARP1 complex by reversible formation of a covalent imine linkage.


Assuntos
Inibidores de Poli(ADP-Ribose) Polimerases , Domínio Catalítico , Gossipol/química , Gossipol/metabolismo , Gossipol/farmacologia , Células HeLa , Humanos , Poli(ADP-Ribose) Polimerases/metabolismo , Análise Serial de Proteínas , Ligação Proteica/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas , Estereoisomerismo
3.
Nat Protoc ; 12(2): 279-288, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28079880

RESUMO

At present, several assays that use radioisotope labeling to quantify the degradation of long-lived proteins have been developed to measure autophagic flux. Here, we describe a nonradioactive pulse-chase protocol using L-azidohomoalanine (AHA) labeling to quantify long-lived protein degradation during autophagy. AHA is used as a surrogate for L-methionine, and, when added to cultured cells grown in methionine-free medium, AHA is incorporated into proteins during de novo protein synthesis. After a chase period to remove short-lived proteins, autophagy is induced by starvation or other stimuli. Cells then undergo a 'click' reaction between the azide group of AHA and a fluorescently tagged alkyne probe. The AHA-containing proteins can then be detected by flow cytometry. This protocol is nonradioactive, sensitive and quantitative, and it is easy to perform. It is also applicable to various cell culture systems. The whole protocol is estimated to take 4-5 d to complete.


Assuntos
Alanina/análogos & derivados , Autofagia , Proteínas/química , Proteínas/metabolismo , Proteólise , Coloração e Rotulagem/métodos , Alanina/química , Alcinos/química , Animais , Azidas/química , Linhagem Celular , Química Click , Corantes Fluorescentes/química , Camundongos
4.
Autophagy ; 12(10): 1931-1944, 2016 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-27463841

RESUMO

Autophagy is an intracellular degradation mechanism in response to nutrient starvation. Via autophagy, some nonessential cellular constituents are degraded in a lysosome-dependent manner to generate biomolecules that can be utilized for maintaining the metabolic homeostasis. Although it is known that under starvation the global protein synthesis is significantly reduced mainly due to suppression of MTOR (mechanistic target of rapamycin serine/threonine kinase), emerging evidence demonstrates that de novo protein synthesis is involved in the autophagic process. However, characterizing these de novo proteins has been an issue with current techniques. Here, we developed a novel method to identify newly synthesized proteins during starvation-mediated autophagy by combining bio-orthogonal noncanonical amino acid tagging (BONCAT) and isobaric tags for relative and absolute quantitation (iTRAQTM). Using bio-orthogonal metabolic tagging, L-azidohomoalanine (AHA) was incorporated into newly synthesized proteins which were then enriched with avidin beads after a click reaction between alkyne-bearing biotin and AHA's bio-orthogonal azide moiety. The enriched proteins were subjected to iTRAQ labeling for protein identification and quantification using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Via the above approach, we identified and quantified a total of 1176 proteins and among them 711 proteins were found to meet our defined criteria as de novo synthesized proteins during starvation-mediated autophagy. The characterized functional profiles of the 711 newly synthesized proteins by bioinformatics analysis suggest their roles in ensuring the prosurvival outcome of autophagy. Finally, we performed validation assays for some selected proteins and found that knockdown of some genes has a significant impact on starvation-induced autophagy. Thus, we think that the BONCAT-iTRAQ approach is effective in the identification of newly synthesized proteins and provides useful insights to the molecular mechanisms and biological functions of autophagy.


Assuntos
Autofagia , Biossíntese de Proteínas , Proteômica/métodos , Alanina/análogos & derivados , Alanina/metabolismo , Ontologia Genética , Células HeLa , Humanos , Reprodutibilidade dos Testes , Transdução de Sinais , Coloração e Rotulagem , Transcrição Gênica
5.
Autophagy ; 11(4): 629-42, 2015 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-25919885

RESUMO

Autophagy is a catabolic process in response to starvation or other stress conditions to sustain cellular homeostasis. At present, histone deacetylase inhibitors (HDACIs) are known to induce autophagy in cells through inhibition of mechanistic target of rapamycin (MTOR) pathway. FOXO1, an important transcription factor regulated by AKT, is also known to play a role in autophagy induction. At present, the role of FOXO1 in the HDACIs-induced autophagy has not been reported. In this study, we first observed that HDACIs increased the expression of FOXO1 at the mRNA and protein level. Second, we found that FOXO1 transcriptional activity was enhanced by HDACIs, as evidenced by increased FOXO1 nuclear accumulation and transcriptional activity. Third, suppression of FOXO1 function by siRNA knockdown or by a chemical inhibitor markedly blocked HDACIs-induced autophagy. Moreover, we found that FOXO1-mediated autophagy is achieved via its transcriptional activation, leading to a dual effect on autophagy induction: (i) enhanced expression of autophagy-related (ATG) genes, and (ii) suppression of MTOR via transcription of the SESN3 (sestrin 3) gene. Finally, we found that inhibition of autophagy markedly enhanced HDACIs-mediated cell death, indicating that autophagy serves as an important cell survival mechanism. Taken together, our studies reveal a novel function of FOXO1 in HDACIs-mediated autophagy in human cancer cells and thus support the development of a novel therapeutic strategy by combining HDACIs and autophagy inhibitors in cancer therapy.


Assuntos
Apoptose/fisiologia , Autofagia/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Inibidores de Histona Desacetilases/metabolismo , Apoptose/genética , Sobrevivência Celular/fisiologia , Proteína Forkhead Box O1 , Humanos , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo
6.
Sci Rep ; 5: 7896, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25600173

RESUMO

Target-identification and understanding of mechanism-of-action (MOA) are challenging for development of small-molecule probes and their application in biology and drug discovery. For example, although aspirin has been widely used for more than 100 years, its molecular targets have not been fully characterized. To cope with this challenge, we developed a novel technique called quantitative acid-cleavable activity-based protein profiling (QA-ABPP) with combination of the following two parts: (i) activity-based protein profiling (ABPP) and iTRAQ™ quantitative proteomics for identification of target proteins and (ii) acid-cleavable linker-based ABPP for identification of peptides with specific binding sites. It is known that reaction of aspirin with its target proteins leads to acetylation. We thus applied the above technique using aspirin-based probes in human cancer HCT116 cells. We identified 1110 target proteins and 2775 peptides with exact acetylation sites. By correlating these two sets of data, 523 proteins were identified as targets of aspirin. We used various biological assays to validate the effects of aspirin on inhibition of protein synthesis and induction of autophagy which were elicited from the pathway analysis of Aspirin target profile. This technique is widely applicable for target identification in the field of drug discovery and biology, especially for the covalent drugs.


Assuntos
Aspirina/química , Peptídeos/química , Proteômica , Acetilação/efeitos dos fármacos , Aspirina/metabolismo , Sítios de Ligação , Descoberta de Drogas , Humanos , Peptídeos/isolamento & purificação , Peptídeos/metabolismo , Análise Serial de Proteínas
7.
Autophagy ; 11(5): 769-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25945613

RESUMO

CAV1 (caveolin 1, caveolae protein, 22kDa) is well known as a principal scaffolding protein of caveolae, a specialized plasma membrane structure. Relatively, the caveolae-independent function of CAV1 is less studied. Autophagy is a process known to involve various membrane structures, including autophagosomes, lysosomes, and autolysosomes for degradation of intracellular proteins and organelles. Currently, the function of CAV1 in autophagy remains largely elusive. In this study, we demonstrate for the first time that CAV1 deficiency promotes both basal and inducible autophagy. Interestingly, the promoting effect was found mainly in the late stage of autophagy via enhancing lysosomal function and autophagosome-lysosome fusion. Notably, the regulatory function of CAV1 in lysosome and autophagy was found to be caveolae-independent, and acts through lipid rafts. Furthermore, the elevated autophagy level induced by CAV1 deficiency serves as a cell survival mechanism under starvation. Importantly, downregulation of CAV1 and enhanced autophagy level were observed in human breast cancer cells and tissues. Taken together, our data reveal a novel function of CAV1 and lipid rafts in breast cancer development via modulation of lysosomal function and autophagy.


Assuntos
Autofagia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caveolina 1/metabolismo , Lisossomos/metabolismo , Estresse Fisiológico , Animais , Caveolina 1/deficiência , Sobrevivência Celular , Regulação para Baixo , Feminino , Humanos , Células MCF-7 , Fusão de Membrana , Microdomínios da Membrana/metabolismo , Camundongos , Modelos Biológicos , Fagossomos/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
8.
Autophagy ; 10(5): 901-12, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24675368

RESUMO

Autophagy is a catabolic process during which cellular components including protein aggregates and organelles are degraded via a lysosome-dependent process to sustain metabolic homeostasis during nutrient or energy deprivation. Measuring the rate of proteolysis of long-lived proteins is a classical assay for measurement of autophagic flux. However, traditional methods, such as a radioisotope labeling assay, are technically tedious and have low sensitivity. Here, we report a novel method for quantification of long-lived protein degradation based on L-azidohomoalanine (AHA) labeling in mouse embryonic fibroblasts (MEFs) and in human cancer cells. AHA is a surrogate for L-methionine, containing a bio-orthogonalazide moiety. When added to cultured cells, AHA is incorporated into proteins during active protein synthesis. After a click reaction between an azide and an alkyne, the azide-containing proteins can be detected with an alkyne-tagged fluorescent dye, coupled with flow cytometry. Induction of autophagy by starvation or mechanistic target of rapamycin (MTOR) inhibitors was able to induce a significant reduction of the fluorescence intensity, consistent with other autophagic markers. Coincidently, inhibition of autophagy by pharmacological agents or by Atg gene deletion abolished the reduction of the fluorescence intensity. Compared with the classical radioisotope pulse-labeling method, we think that our method is sensitive, quantitative, nonradioactive, and easy to perform, and can be applied to both human and animal cell culture systems.


Assuntos
Alanina/análogos & derivados , Autofagia , Bioensaio/métodos , Proteólise , Coloração e Rotulagem/métodos , Alanina/química , Alanina/farmacologia , Animais , Sobrevivência Celular , Células Cultivadas , Citometria de Fluxo/métodos , Corantes Fluorescentes/análise , Corantes Fluorescentes/metabolismo , Células Hep G2 , Humanos , Camundongos
9.
FEBS Lett ; 587(2): 170-7, 2013 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-23201261

RESUMO

In this study we aim to elucidate the signaling pathway and biological function of autophagy induced by MNNG, a commonly used DNA alkylating agent. We first observed that MNNG is able to induce necrotic cell death and autophagy in Bax-/- Bak-/- double knockout MEFs. We analyzed the critical role of PARP-1 activation and ATP depletion in MNNG-mediated cell death and autophagy via AMPK activation and mTOR suppression. We provide evidence that suppression of AMPK blocks MNNG-induced autophagy and enhances cell death, suggesting the pro-survival function of autophagy in MNNG-treated cells. Taken together, data from this study reveal a novel mechanism in controlling MNNG-mediated autophagy via AMPK activation downstream of PARP-1 activation and ATP depletion.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Alquilantes/farmacologia , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Técnicas de Inativação de Genes , Metilnitronitrosoguanidina/farmacologia , Camundongos , Modelos Biológicos , Poli(ADP-Ribose) Polimerase-1 , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/genética , RNA Interferente Pequeno/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2/deficiência , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína X Associada a bcl-2/deficiência , Proteína X Associada a bcl-2/genética
10.
Autophagy ; 7(10): 1173-86, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21808151

RESUMO

It has been well documented that cells deficient in either TSC1 or TSC2 are highly sensitive to various cell death stimuli. In this study, we utilized the TSC2 (-/-) mouse embryonic fibroblasts (MEFs) to study the involvement of autophagy in the enhanced susceptibility of TSC2-null cells to cell death. We first confirmed that both TSC1-null and TSC2-null MEFs are more sensitive to apoptosis in response to amino acid starvation (EBSS) and hypoxia. Second, we found that both the basal and inducible autophagy in TSC2 (-/-) MEFs is impaired, mainly due to constitutive activation of mTORC1. Third, suppression of autophagy by chloroquine and Atg7 knockdown sensitizes TSC2 (+/+) cells, but not TSC2 (-/-) cells, to EBSS-induced cell death. Conversely, the inhibition of mTORC1 by raptor knockdown and rapamycin activates autophagy and subsequently rescues TSC2 (-/-) cells. Finally, in starved cells, nutrient supplementations (insulin-like growth factor-1 (IGF-1) and leucine) enhanced cell death in TSC2 (-/-) cells, but reduced cell death in TSC2 (+/+) cells. Taken together, these data indicate that constitutive activation of mTORC1 in TSC2 (-/-) cells leads to suppression of autophagy and enhanced susceptibility to stress-mediated cell death. Our findings thus provide new insights into the complex relationships among mTOR, autophagy and cell death, and support the possible autophagy-targeted intervention strategies for the treatment of TSC-related pathologies.


Assuntos
Autofagia , Serina-Treonina Quinases TOR/metabolismo , Animais , Proteína 7 Relacionada à Autofagia , Morte Celular , Linhagem Celular , Sobrevivência Celular , Cloroquina/farmacologia , Fibroblastos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Complexos Multiproteicos , Proteínas/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fatores de Tempo , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA