Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Blood Adv ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38861356

RESUMO

ß-thalassemia is a condition characterized by reduced or absent synthesis of ß-globin resulting from genetic mutations, leading to expanded and ineffective erythropoiesis. Mitoxantrone has been widely used clinically as an antitumor agent in light of its ability to inhibit cell proliferation. However, its therapeutic effect on expanded and ineffective erythropoiesis in ß-thalassemia is untested. We found that mitoxantrone decreased α-globin precipitates and ameliorated anemia, splenomegaly and ineffective erythropoiesis in the HbbTh3/+ mouse model of ß-thalassemia intermedia. The partially reversed ineffective erythropoiesis is a consequence of effects on autophagy as mitochondrial retention and protein levels of mTOR, P62 and LC3 in reticulocytes decreased in mitoxantrone-treated HbbTh3/+ mice. These data provide significant pre-clinical evidence for targeting autophagy as a novel therapeutic approach for ß-thalassemia.

2.
Pharmacol Res ; 194: 106835, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37348691

RESUMO

Maintaining mitochondrial homeostasis is a potential therapeutic strategy for various diseases, including neurodegenerative diseases, cardiovascular diseases, metabolic disorders, and cancer. Selective degradation of mitochondria by autophagy (mitophagy) is a fundamental mitochondrial quality control mechanism conserved from yeast to humans. Indeed, small-molecule modulators of mitophagy are valuable pharmaceutical tools that can be used to dissect complex biological processes and turn them into potential drugs. In the past few years, pharmacological regulation of mitophagy has shown promising therapeutic efficacy in various disease models. However, with the increasing number of chemical mitophagy modulator studies, frequent methodological flaws can be observed, leading some studies to draw unreliable or misleading conclusions. This review attempts (a) to summarize the molecular mechanisms of mitophagy; (b) to propose a Mitophagy Modulator Characterization System (MMCS); (c) to perform a comprehensive analysis of methods used to characterize mitophagy modulators, covering publications over the past 20 years; (d) to provide novel targets for pharmacological intervention of mitophagy. We believe this review will provide a panorama of current research on chemical mitophagy modulators and promote the development of safe and robust mitophagy modulators with therapeutic potential by introducing high methodological standards.


Assuntos
Doenças Cardiovasculares , Neoplasias , Humanos , Mitofagia , Autofagia , Mitocôndrias/metabolismo , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
3.
Hepatology ; 76(5): 1389-1408, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35313046

RESUMO

BACKGROUND AND AIMS: Sam50, a key component of the sorting and assembly machinery (SAM) complex, is also involved in bridging mitochondrial outer-membrane and inner-membrane contacts. However, the physiological and pathological functions of Sam50 remain largely unknown. APPROACH AND RESULTS: Here we show that Sam50 interacts with MICOS (mitochondrial contact site and cristae organizing system) and ATAD3 (ATPase family AAA domain-containing protein 3) to form the Sam50-MICOS-ATAD3-mtDNA axis, which maintains mtDNA stability. Loss of Sam50 causes mitochondrial DNA (mtDNA) aggregation. Furthermore, Sam50 cooperates with Mic60 to bind to cardiolipin, maintaining the integrity of mitochondrial membranes. Sam50 depletion leads to cardiolipin externalization, which causes mitochondrial outer-membrane and inner-membrane (including crista membrane) remodeling, triggering Bax mitochondrial recruitment, mtDNA aggregation, and release. Physiologically, acetaminophen (an effective antipyretic and analgesic)-caused Sam50 reduction or Sam50 liver-specific knockout induces mtDNA release, leading to activation of the cGAS-STING pathway and liver inflammation in mice. Moreover, exogenous expression of Sam50 remarkably attenuates APAP-induced liver hepatoxicity. CONCLUSIONS: Our findings uncover the critical role of Sam50 in maintaining mitochondrial membrane integrity and mtDNA stability in hepatocytes and reveal that Sam50 depletion-induced cardiolipin externalization is a signal of mtDNA release and controls mtDNA-dependent innate immunity.


Assuntos
Antipiréticos , Membranas Mitocondriais , Animais , Humanos , Camundongos , Acetaminofen , Adenosina Trifosfatases/metabolismo , Proteína X Associada a bcl-2/metabolismo , Cardiolipinas/metabolismo , DNA Mitocondrial/genética , Células HeLa , Hepatócitos/metabolismo , Fígado/metabolismo , Proteínas de Membrana/genética , Membranas Mitocondriais/metabolismo , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Proteínas Mitocondriais/metabolismo , Nucleotidiltransferases/metabolismo
4.
Cell Death Dis ; 12(9): 837, 2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34489398

RESUMO

Mitochondrial retrograde signaling (mito-RTG) triggered by mitochondrial dysfunction plays a potential role in regulating tumor metabolic reprogramming and cellular sensitivity to radiation. Our previous studies showed phos-pyruvate dehydrogenase (p-PDH) and PDK1, which involved in aerobic glycolysis, were positively correlated with radioresistance, but how they initiate and work in the mito-RTG pathway is still unknown. Our further genomics analysis revealed that complex I components were widely downregulated in mitochondrial dysfunction model. In the present study, high expression of p-PDH was found in the complex I deficient cells and induced radioresistance. Mechanistically, complex I defects led to a decreased PDH both in cytoplasm and nucleus through [Ca2+]m-PDP1-PDH axis, and decreased PDH in nucleus promote DNA damage repair (DDR) response via reducing histone acetylation. Meanwhile, NDUFS1 (an important component of the complex I) overexpression could enhance the complex I activity, reverse glycolysis and resensitize cancer cells to radiation in vivo and in vitro. Furthermore, low NDUFS1 and PDH expression were validated to be correlated with poor tumor regression grading (TRG) in local advanced colorectal cancer (CRC) patients underwent neoadjuvant radiotherapy. Here, we propose that the [Ca2+]m-PDP1-PDH-histone acetylation retrograde signaling activated by mitochondrial complex I defects contribute to cancer cell radioresistance, which provides new insight in the understanding of the mito-RTG. For the first time, we reveal that NDUFS1 could be served as a promising predictor of radiosensitivity and modification of complex I function may improve clinical benefits of radiotherapy in CRC.


Assuntos
Cálcio/metabolismo , Neoplasias Colorretais/metabolismo , Histonas/metabolismo , Mitocôndrias/patologia , Proteína Fosfatase 2C/metabolismo , Complexo Piruvato Desidrogenase/metabolismo , Tolerância a Radiação , Transdução de Sinais , Acetilação , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Dano ao DNA , Reparo do DNA , Intervalo Livre de Doença , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Glicólise , Humanos , Estimativa de Kaplan-Meier , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , NADH Desidrogenase/metabolismo , Rotenona/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Adv Sci (Weinh) ; 8(19): e2100779, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34363353

RESUMO

In eukaryote cells, core components of chromatin, such as histones and DNA, are packaged in nucleus. Leakage of nuclear materials into cytosol will induce pathological effects. However, the underlying mechanisms remain elusive. Here, cytoplasmic localization of nuclear materials induced by chromatin dysregulation (CLIC) in mammalian cells is reported. H3K9me3 inhibition by small chemicals, HP1α knockdown, or knockout of H3K9 methylase SETDB1, induces formation of cytoplasmic puncta containing histones H3.1, H4 and cytosolic DNA, which in turn activates inflammatory genes and autophagic degradation. Autophagy deficiency rescues H3 degradation, and enhances the activation of inflammatory genes. MRE11, a subunit of MRN complex, enters cytoplasm after heterochromatin dysregulation. Deficiency of MRE11 or NBS1, but not RAD50, inhibits CLIC puncta in cytosol. MRE11 depletion represses tumor growth enhanced by HP1α deficiency, suggesting a connection between CLIC and tumorigenesis. This study reveals a novel pathway that heterochromatin dysregulation induces translocation of nuclear materials into cytoplasm, which is important for inflammatory diseases and cancer.


Assuntos
Citoplasma/genética , Citoplasma/metabolismo , Epigênese Genética/genética , Histonas/genética , Histonas/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Fatores de Transcrição/genética
6.
EMBO Rep ; 22(1): e50827, 2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33314701

RESUMO

Many cancer cells maintain enhanced aerobic glycolysis due to irreversible defective mitochondrial oxidative phosphorylation (OXPHOS). This phenomenon, known as the Warburg effect, is recently challenged because most cancer cells maintain OXPHOS. However, how cancer cells coordinate glycolysis and OXPHOS remains largely unknown. Here, we demonstrate that OMA1, a stress-activated mitochondrial protease, promotes colorectal cancer development by driving metabolic reprogramming. OMA1 knockout suppresses colorectal cancer development in AOM/DSS and xenograft mice models of colorectal cancer. OMA1-OPA1 axis is activated by hypoxia, increasing mitochondrial ROS to stabilize HIF-1α, thereby promoting glycolysis in colorectal cancer cells. On the other hand, under hypoxia, OMA1 depletion promotes accumulation of NDUFB5, NDUFB6, NDUFA4, and COX4L1, supporting that OMA1 suppresses OXPHOS in colorectal cancer. Therefore, our findings support a role for OMA1 in coordination of glycolysis and OXPHOS to promote colorectal cancer development and highlight OMA1 as a potential target for colorectal cancer therapy.


Assuntos
Neoplasias Colorretais , Fosforilação Oxidativa , Animais , Ciclo do Ácido Cítrico , Neoplasias Colorretais/genética , Glicólise , Hipóxia/genética , Camundongos
7.
Front Cell Dev Biol ; 8: 180, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32373606

RESUMO

Optic Atrophy 1 (OPA1) has well-established roles in both mitochondrial fusion and apoptotic crista remodeling and is required for the maintenance and distribution of mitochondrial DNA (mtDNA), which are essential for energy metabolism. However, the relationship between OPA1 and mitochondrial metabolism and the underlying mechanisms remain unclear. Here, we show that OPA1-Exon4b modulates mitochondrial respiration and rescues inner mitochondrial membrane potential (Δψm), independent of mitochondrial fusion. OPA1-Exon4b is required for the maintenance of normal TFAM distribution and enhances mtDNA transcription by binding the D-loop of mtDNA. Finally, we show that mRNA levels of OPA1 isoforms containing Exon4b are specifically downregulated in hepatocellular carcinoma (HCC), leading to a reduction in Δψm. Thus, our study demonstrates a novel mitochondrial functional self-recovery pathway involving enhanced mtDNA transcription-mediated recovery of mitochondrial respiratory chain proteins. This mitochondrial fusion-independent pathway may contribute to mitochondrial multi-functional switches in tumorigenesis.

8.
Autophagy ; 16(3): 419-434, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31177901

RESUMO

Mitophagy, which is a conserved cellular process for selectively removing damaged or unwanted mitochondria, is critical for mitochondrial quality control and the maintenance of normal cellular physiology. However, the precise mechanisms underlying mitophagy remain largely unknown. Prior studies on mitophagy focused on the events in the mitochondrial outer membrane. PHB2 (prohibitin 2), which is a highly conserved membrane scaffold protein, was recently identified as a novel inner membrane mitophagy receptor that mediates mitophagy. Here, we report a new signaling pathway for PHB2-mediated mitophagy. Upon mitochondrial membrane depolarization or misfolded protein aggregation, PHB2 depletion destabilizes PINK1 in the mitochondria, which blocks the mitochondrial recruitment of PRKN/Parkin, ubiquitin and OPTN (optineurin), leading to an inhibition of mitophagy. In addition, PHB2 overexpression directly induces PRKN recruitment to the mitochondria. Moreover, PHB2-mediated mitophagy is dependent on the mitochondrial inner membrane protease PARL, which interacts with PHB2 and is activated upon PHB2 depletion. Furthermore, PGAM5, which is processed by PARL, participates in PHB2-mediated PINK1 stabilization. Finally, a ligand of PHB proteins that we synthesized, called FL3, was found to strongly inhibit PHB2-mediated mitophagy and to effectively block cancer cell growth and energy production at nanomolar concentrations. Thus, our findings reveal that the PHB2-PARL-PGAM5-PINK1 axis is a novel pathway of PHB2-mediated mitophagy and that targeting PHB2 with the chemical compound FL3 is a promising strategy for cancer therapy.Abbreviations: AIFM1: apoptosis inducing factor mitochondria associated 1; ATP5F1A/ATP5A1: ATP synthase F1 subunit alpha; BAF: bafilomycin A1; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CCCP: chemical reagent carbonyl cyanide m-chlorophenyl hydrazine; FL3: flavaglines compound 3; HSPD1/HSP60: heat shock protein family D (Hsp60) member 1; LC3B/MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MEF: mouse embryo fibroblasts; MPP: mitochondrial-processing peptidase; MT-CO2/COX2: mitochondrially encoded cytochrome c oxidase II; MTS: mitochondrial targeting sequence; OA: oligomycin and antimycin A; OPTN: optineurin; OTC: ornithine carbamoyltransferase; PARL: presenilin associated rhomboid like; PBS: phosphate-buffered saline; PGAM5: PGAM family member 5, mitochondrial serine/threonine protein phosphatase; PHB: prohibitin; PHB2: prohibitin 2; PINK1: PTEN induced kinase 1; PRKN/Parkin: parkin RBR E3 ubiquitin protein ligase; Roc-A: rocaglamide A; TOMM20: translocase of outer mitochondrial membrane 20; TUBB: tubulin beta class I.


Assuntos
Metaloproteases/metabolismo , Proteínas Mitocondriais/metabolismo , Mitofagia , Fosfoproteínas Fosfatases/metabolismo , Proteínas Quinases/metabolismo , Proteínas Repressoras/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Células HCT116 , Células HeLa , Humanos , Ligantes , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Proibitinas , Ligação Proteica , Estabilidade Proteica , Transdução de Sinais
9.
EMBO J ; 36(23): 3483-3500, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29046333

RESUMO

The tumor suppressor p53 is activated in response to cellular stress to prevent malignant transformation. However, several recent studies have shown that p53 can play protective roles in tumor cell survival under adversity. Whether p53-regulated long noncoding RNAs are involved in this process remains to be fully understood. Here, we show that under glucose starvation condition, p53 directly upregulates a novel lncRNA named TRINGS (Tp53-regulated inhibitor of necrosis under glucose starvation) in human tumor cells. TRINGS binds to STRAP and inhibits STRAP-GSK3ß-NF-κB necrotic signaling to protect tumor cells from cell death. Interestingly, TRINGS appears to respond to glucose starvation specifically, as it is not activated by serum, serine, or glutamine deprivation. Collectively, our findings reveal that p53-induced lncRNA TRINGS controls the necrotic pathway and contributes to the survival of cancer cells harboring wild-type p53 under glucose stress.


Assuntos
Genes p53 , Neoplasias/genética , Neoplasias/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , NF-kappa B/metabolismo , Necrose , Proteínas de Neoplasias/metabolismo , Neoplasias/patologia , Ligação Proteica , RNA Longo não Codificante/antagonistas & inibidores , RNA Neoplásico/antagonistas & inibidores , Proteínas de Ligação a RNA , Regulação para Cima
10.
Sci Rep ; 5: 16064, 2015 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-26530328

RESUMO

The inner mitochondrial membrane (IMM) invaginates to form cristae and the maintenance of cristae depends on the mitochondrial contact site (MICOS) complex. Mitofilin and CHCHD6, which physically interact, are two components of the MICOS. In this study, we performed immunoprecipitation experiments with Mitofilin and CHCHD6 antibodies and identified a complex containing Mitofilin, Sam50, and CHCHD 3 and 6. Using transcription activator-like effector nucleases (TALENs), we generated knockdown/knockout clones of Mitofilin and CHCHD6. Transmission electron microscopy (TEM) revealed that vesicle-like cristae morphology appeared in cell lines lacking Mitofilin, and mitochondria exhibited lower cristae density in CHCHD6-knockout cells. Immunoblot analysis showed that knockdown of Mitofilin, but not knockout of CHCHD6, affected their binding partners that control cristae morphology. We also demonstrated that Mitofilin and CHCHD6 directly interacted with Sam50. Additionally, we observed that Mitofilin-knockdown cells showed decreased mitochondrial membrane potential (ΔΨm) and intracellular ATP content, which were minimally affected in CHCHD6-knockout cells. Taken together, we conclude that the integrity of MICOS and its efficient interaction with Sam50 are indispensable for cristae organization, which is relevant to mitochondrial function.


Assuntos
Potencial da Membrana Mitocondrial/genética , Proteínas de Membrana/metabolismo , Membranas Mitocondriais/fisiologia , Proteínas Mitocondriais/metabolismo , Proteínas Musculares/metabolismo , Trifosfato de Adenosina/metabolismo , Anticorpos/imunologia , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Microscopia Eletrônica de Transmissão , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/imunologia , Complexos Multiproteicos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/imunologia
11.
Oncogene ; 24(16): 2723-34, 2005 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-15735764

RESUMO

For a long time, as the most prominent subnuclear structure, nucleolus has been recognized as a main site where rRNA processing and ribosomal subunit assemblies take place. It has not been until recently that additional functions of nucleolus have begun to be proposed. In this study, we for the first time demonstrate that Survivin-deltaEx3, a novel functionally splice variant of Survivin localizes in the nucleoli where it degrades rapidly through ubiquitin-proteosome pathway. Several lines of evidences provided in this report support this finding (i) a novel nucleolar localization sequence (NoLS, MQRKPTIRRKNLRLRRK) and a novel degradation signal (aa92-aa137) within Survivin-deltaEx3 were identified (ii) proteasome inhibitors MG132 or ALLN greatly inhibits degradation of Survivin-deltaEx3 and polyubiquitination of Survivin-deltaEx3 was detected (iii) heterologous proteins such as TAT-PTD or p14ARF, when fused to this putative degradation signal, result in a significant degradation within the nucleolus. In addition, the nucleolar localization and degradation of Survivin-deltaEx3 appear to be required for its antiapoptotic function, since neither NoLS-deleted nor degradation signal-deleted Survivin-deltaEx3 retains protective effect against Doxorubicin-induced apoptosis. Thus, our results have provided evidences to suggest that besides cytosol, nucleus, endoplsmic reticulum (ER) or lysosomes, nucleolus may also operate important protein degradation pathway, which has been overlooked previously.


Assuntos
Nucléolo Celular/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Animais , Apoptose , Compartimento Celular , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose , Camundongos , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/genética , Proteínas de Neoplasias , Oligonucleotídeos/genética , Plasmídeos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Survivina , Transfecção , Células Tumorais Cultivadas
12.
J Pharmacol Exp Ther ; 311(1): 22-33, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15173313

RESUMO

We have studied the effect of tri-phenyl tin benzimadazolethiolcopper chloride (TPT-CuCl(2)), a novel bimetallic compound, on the regulation of apoptosis in HeLa cells, MCF-7 cells, and in vivo Wistar rat model. TPT-CuCl(2) induces significant apoptosis in HeLa cell line characterized by DNA fragmentation and chromosome condensation. Comet assay revealed that TPT-CuCl(2) targets and causes severe damage to the DNA. Treatment of HeLa cells with TPT-CuCl(2) rescues the accumulation of p53 from the suppression of human papilloma virus E6, resulting in a dramatic up-regulation of Bax and Bak and down-regulation of the antiapoptotic factor Survivin. Apoptotic induction by TPT-CuCl(2) was shown to mediate in a p53-depedent manner; loss of p53 impairs the release of cytochrome c and Smac/DIABLO from mitochondria to cytosol. Moreover, we have shown that TPT-CuCl(2) induced-apoptosis was through an intrinsic mitochondrial pathway, which was inhibited by viral oncoprotein E1B19K. Caspase-3 was found to be indispensable in TPT-CuCl(2)-triggered apoptosis signaling pathway, because caspase-3 deficient cell line MCF-7 was resistant to TPT-CuCl(2). Furthermore, in vivo studies using C6 glioblastoma xenograft rat model revealed that TPT-CuCl(2) exhibits significant antiproliferative activity against tumor development with minimal cytotoxicity toward normal physiological function of the experimental rats. These findings imply the attractiveness of TPT-CuCl(2) as a drug candidate for further development.


Assuntos
Apoptose , Compostos Orgânicos de Estanho/farmacologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/metabolismo , Caspase 3 , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Citocromos c/metabolismo , Regulação para Baixo , Ativação Enzimática , Feminino , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas de Neoplasias , Papillomaviridae/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Survivina , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/patologia , Proteína Killer-Antagonista Homóloga a bcl-2 , Proteína X Associada a bcl-2
13.
Mol Biol Cell ; 15(3): 1287-96, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14699067

RESUMO

Survivin is a member of the inhibitor of apoptosis protein (IAP) family that has been implicated in both apoptosis inhibition and cell cycle control. Recently, Survivin has attracted growing attention because of its tumor-specific expression and potential applications in tumor therapy. However, its inhibitory mechanism and subcellular localization remain controversial. Here, we report a novel Survivin mutant Surv-D53A, which displays a function opposite to Survivin and a distinctive subcellular distribution compared with its wild-type counterpart. Surv-D53A was shown to induce apoptosis in a p53-independent manner, indicating that tumor suppressor p53 is not involved in its apoptosis pathway. Surv-D53A was shown to markedly sensitize apoptosis induced by TRAIL, doxorubicin, and RIP3. We also demonstrated that similar to wild-type Survivin, Surv-D53A was localized in cytoplasm in interphase and to midbody at telophase. However, it fails to colocalize in chromosomes with Aurora-B in metaphase as wt-Survivin. Surv-D53A mutant is less stable than wt-Survivin and is degraded more rapidly by ubiquitin-proteasome pathway. Additionally, we found that Surv-D53A interacts with wt-Survivin to form heterodimer or with itself to form mutant homodimer, which may account for the loss of its antiapoptotic function. Finally, unlike Survivin*Survivin, neither Surv-D53A*Survivin nor Surv-D53A*Surv-D53A is able to bind to Smac/DIABLO, which may explain the underlying mechanism for its abolishment of antiapoptotic activity of Survivin.


Assuntos
Substituição de Aminoácidos/genética , Apoptose/fisiologia , Sobrevivência Celular/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Aurora Quinase B , Aurora Quinases , Proteínas de Transporte/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Cromossomos/metabolismo , Citoplasma/metabolismo , Dimerização , Doxorrubicina/farmacologia , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana/farmacologia , Proteínas Mitocondriais/metabolismo , Dados de Sequência Molecular , Proteínas de Neoplasias , Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores , Survivina , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia
14.
J Biol Chem ; 278(25): 23130-40, 2003 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12660240

RESUMO

Survivin is a member of the inhibitor of apoptosis protein (IAP) family that has been implicated in both apoptosis inhibition and cell cycle control. However, its inhibitory mechanism and subcellular localization remain controversial. In this report, we provided evidence for the first time that Survivin physically interacts with Smac/DIABLO both in vitro and in vivo. A point mutation (D71R) in the baculovirus IAP repeat motif and a C-terminal deletion mutant (Surv-BIR) of Survivin fail to bind to Smac/DIABLO and abrogate its ability to inhibit apoptosis. The N-terminal of mature Smac/DIABLO is absolutely required for Survivin.Smac complex formation. Subcellular distributions of Survivin and Smac/DIABLO showed that they co-localized within the cytosol during interphase. In addition, Survivin was found to be incapable of binding to caspase. We also identified that the co-presence of Smac/DIABLO and XIAP was required for Survivin to inhibit caspase cleavage in a cell-free system. In conclusion, our results provide the first evidence that the interaction between Smac/DIABLO and Survivin is an essential step underling the inhibition of apoptosis induced by Taxol.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Transporte/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas Mitocondriais/fisiologia , Paclitaxel/farmacologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Antígenos de Neoplasias/farmacologia , Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose , Sítios de Ligação , Ciclo Celular/fisiologia , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Associadas aos Microtúbulos/genética , Mitocôndrias/fisiologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas de Neoplasias , Mutação Puntual , Reação em Cadeia da Polimerase , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes/farmacologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Survivina , Transfecção
15.
FEBS Lett ; 532(1-2): 36-44, 2002 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-12459459

RESUMO

Several novel prokaryotic and eukaryotic expression vectors were constructed for protein transduction and subcellular localization. These vectors employed an N-terminal stretch of 11 basic amino acid residues (47-57) from the human immunodeficiency virus type 1 (HIV-1) TAT protein transduction domain (PTD) for protein translocation and cellular localization. The vectors also contained a six-histidine (His(6)) tag at the N- or C-terminus for convenient purification and detection, and a multiple cloning site for easy insertion of foreign genes. Some heterologous genes including HSV-TK, Bcl-rambo, Smac/DIABLO and GFP were fused in-frame to TAT PTD and successfully overexpressed in Escherichia coli. The purified TAT-GFP fusion protein was able to transduce into the mammalian cells and was found to locate mainly in the cytosol when exogenously added to the cell culture medium. However, using a transfection system, mammalian-expressed TAT-GFP predominantly displayed a nuclear localization and nucleolar accumulation in mammalian cell lines. This discrepancy implies that the exact subcellular localization of transduced protein may depend on cell type, the nature of imported proteins and delivery approach. Taken together, our results demonstrate that a TAT PTD length of 11 amino acids was sufficient to confer protein internalization and its subsequent cellular localization. These novel properties allow these vectors to be useful for studying protein transduction and nuclear import.


Assuntos
Produtos do Gene tat/genética , Vetores Genéticos , HIV-1 , Transporte Ativo do Núcleo Celular , Animais , Proteínas Reguladoras de Apoptose , Sequência de Bases , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Núcleo Celular/química , Núcleo Celular/metabolismo , Citosol/química , Escherichia coli/genética , Produtos do Gene tat/análise , Produtos do Gene tat/química , Produtos do Gene tat/metabolismo , Vetores Genéticos/química , Proteínas de Fluorescência Verde , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Dados de Sequência Molecular , Sinais de Localização Nuclear , Peptídeos/farmacologia , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/metabolismo , Células Tumorais Cultivadas , Produtos do Gene tat do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA