Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 7(24)2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36413415

RESUMO

Metastatic clear cell renal cell carcinomas (ccRCCs) are resistant to DNA-damaging chemotherapies, limiting therapeutic options for patients whose tumors are resistant to tyrosine kinase inhibitors and/or immune checkpoint therapies. Here we show that mouse and human ccRCCs were frequently characterized by high levels of endogenous DNA damage and that cultured ccRCC cells exhibited intact cellular responses to chemotherapy-induced DNA damage. We identify that pharmacological inhibition of the DNA damage-sensing kinase ataxia telangiectasia and Rad3-related protein (ATR) with the orally administered, potent, and selective drug M4344 (gartisertib) induced antiproliferative effects in ccRCC cells. This effect was due to replication stress and accumulation of DNA damage in S phase. In some cells, DNA damage persisted into subsequent G2/M and G1 phases, leading to the frequent accumulation of micronuclei. Daily single-agent treatment with M4344 inhibited the growth of ccRCC xenograft tumors. M4344 synergized with chemotherapeutic drugs including cisplatin and carboplatin and the poly(ADP-ribose) polymerase inhibitor olaparib in mouse and human ccRCC cells. Weekly M4344 plus cisplatin treatment showed therapeutic synergy in ccRCC xenografts and was efficacious in an autochthonous mouse ccRCC model. These studies identify ATR inhibition as a potential novel therapeutic option for ccRCC.


Assuntos
Antineoplásicos , Carcinoma de Células Renais , Humanos , Animais , Camundongos , Carcinoma de Células Renais/tratamento farmacológico , Cisplatino , Proteínas Mutadas de Ataxia Telangiectasia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico
2.
Oncotarget ; 11(48): 4490-4503, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-33400734

RESUMO

Tumor cells typically enhance their metabolic capacity to sustain their higher rate of growth and proliferation. One way to elevate the nutrient intake into cancer cells is to increase the expression of genes encoding amino acid transporters, which may represent targetable vulnerabilities. Here, we study the regulation and function of the broad amino acid transporter SLC6A14 in combination with metabolic stress, providing insights into an uncharacterized aspect of the transporter activity. We analyze the pattern of transcriptional changes in a panel of breast cancer cell lines upon metabolic stress and found that SLC6A14 expression levels are increased in the absence of methionine. Methionine deprivation, which can be achieved via modulation of dietary methionine intake in tumor cells, in turn leads to a heightened activation of the AMP-activated kinase (AMPK) in SLC6A14-deficient cells. While SLC6A14 genetic deficiency does not have a major impact on cell proliferation, combined depletion of AMPK and SLC6A14 leads to an increase in apoptosis upon methionine starvation, suggesting that combined targeting of SLC6A14 and AMPK can be exploited as a therapeutic approach to starve tumor cells.

3.
Commun Biol ; 1: 210, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30511023

RESUMO

Cytotoxic activities of several Golgi-dispersing compounds including AMF-26/M-COPA, brefeldin A and golgicide A have previously been shown to induce autophagy or apoptosis. Here, we demonstrate that these Golgi disruptors also trigger ferroptosis, a non-apoptotic form of cell death characterized by iron-dependent oxidative degradation of lipids. Inhibitors of ferroptosis not only counteract cell death, but they also protect from Golgi dispersal and inhibition of protein secretion in response to several Golgi stress agents. Furthermore, the application of sublethal doses of ferroptosis-inducers such as erastin and sorafenib, low cystine growth conditions, or genetic knockdown of SLC7A11 and GPX4 all similarly protect cells from Golgi stress and lead to modulation of ACSL4, SLC7A5, SLC7A11 or GPX4 levels. Collectively, this study suggests a previously unrecognized function of the Golgi apparatus, which involves cellular redox control and prevents ferroptotic cell death.

4.
Mol Biol Cell ; 29(1): 42-52, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29118074

RESUMO

The secretory pathway is a major determinant of cellular homoeostasis. While research into secretory stress signaling has so far mostly focused on the endoplasmic reticulum (ER), emerging data suggest that the Golgi itself serves as an important signaling hub capable of initiating stress responses. To systematically identify novel Golgi stress mediators, we performed a transcriptomic analysis of cells exposed to three different pharmacological compounds known to elicit Golgi fragmentation: brefeldin A, golgicide A, and monensin. Subsequent gene-set enrichment analysis revealed a significant contribution of the ETS family transcription factors ELK1, GABPA/B, and ETS1 to the control of gene expression following compound treatment. Induction of Golgi stress leads to a late activation of the ETS upstream kinases MEK1/2 and ERK1/2, resulting in enhanced ETS factor activity and the transcription of ETS family target genes related to spliceosome function and cell death induction via alternate MCL1 splicing. Further genetic analyses using loss-of-function and gain-of-function experiments suggest that these transcription factors operate in parallel.


Assuntos
Processamento Alternativo/genética , Complexo de Golgi/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Estresse Fisiológico , Transcrição Gênica , Células A549 , Processamento Alternativo/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Brefeldina A/farmacologia , Citoproteção/efeitos dos fármacos , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Complexo de Golgi/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monensin/farmacologia , Piridinas/farmacologia , Quinolinas/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Spliceossomos/efeitos dos fármacos , Spliceossomos/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Regulação para Cima/efeitos dos fármacos
5.
Cell Death Dis ; 8(10): e3069, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28981087

RESUMO

Disruption of the Golgi apparatus can induce a distinct form of programmed cell death that has not been thoroughly characterized. We found that pharmacological application of Golgi stress leads to induction of death receptors (DRs) 4 and 5. DR4 appears to be primarily responsible for the initiation of cell death downstream of Golgi stress, whereas DR5 seems to be more important for cell death triggered by endoplasmic reticulum (ER) stress in specific cancer cell lines. DR induction downstream of either Golgi or ER stress mainly causes intracellular accumulation of DR4 presumably at the Golgi, rather than increased expression on the cell surface. Nevertheless, cells treated with secretory pathway stressors displayed an increased susceptibility to TRAIL (tumor necrosis factor related apoptosis inducing ligand), the endogenous ligand of DR4/5, probably due to intracellular sequestration of the caspase-8 regulator CFLAR (caspase-8 and FADD-like apoptosis regulator). These findings have implications for the treatment of cancer with DR agonists and our general understanding of DR signaling while highlighting the role of the Golgi apparatus as a cell death signaling platform.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Apoptose/efeitos dos fármacos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Caspase 8/genética , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/genética , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/genética , Humanos , Neoplasias/genética , Neoplasias/patologia
6.
Mol Biol Cell ; 28(26): 3756-3772, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29074567

RESUMO

The Golgi apparatus is increasingly recognized as a major hub for cellular signaling and is involved in numerous pathologies, including neurodegenerative diseases and cancer. The study of Golgi stress-induced signaling pathways relies on the selectivity of the available tool compounds of which currently only a few are known. To discover novel Golgi-fragmenting agents, transcriptomic profiles of cells treated with brefeldin A, golgicide A, or monensin were generated and compared with a database of gene expression profiles from cells treated with other bioactive small molecules. In parallel, a phenotypic screen was performed for compounds that alter normal Golgi structure. Histone deacetylase (HDAC) inhibitors and DNA-damaging agents were identified as novel Golgi disruptors. Further analysis identified HDAC1/HDAC9 as well as BRD8 and DNA-PK as important regulators of Golgi breakdown mediated by HDAC inhibition. We provide evidence that combinatorial HDACi/(+)-JQ1 treatment spurs synergistic Golgi dispersal in several cancer cell lines, pinpointing a possible link between drug-induced toxicity and Golgi morphology alterations.


Assuntos
Azepinas/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Histona Desacetilase 1/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Triazóis/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Perfilação da Expressão Gênica/métodos , Complexo de Golgi/efeitos dos fármacos , Histona Desacetilase 1/genética , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos
7.
Traffic ; 18(8): 530-544, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28485883

RESUMO

The Golgi apparatus is part of the secretory pathway and of central importance for modification, transport and sorting of proteins and lipids. ADP-ribosylation factors, whose activation can be blocked by brefeldin A (BFA), play a major role in functioning of the Golgi network and regulation of membrane traffic and are also involved in proliferation and migration of cancer cells. Due to high cytotoxicity and poor bioavailability, BFA has not passed the preclinical stage of drug development. Recently, AMF-26 and golgicide A have been described as novel inhibitors of the Golgi system with antitumor or bactericidal properties. We provide here further evidence that AMF-26 closely mirrors the mode of action of BFA but is less potent. Using several human cancer cell lines, we studied the effects of AMF-26, BFA and golgicide A on cell homeostasis including Golgi structure, endoplasmic reticulum (ER) stress markers, secretion and viability, and found overall a significant correlation between these parameters. Furthermore, modulation of ADP-ribosylation factor expression has a profound impact on Golgi organization and survival in response to Golgi stress inducers.


Assuntos
Sobrevivência Celular , Complexo de Golgi/metabolismo , Estresse Fisiológico , Fatores de Ribosilação do ADP/efeitos dos fármacos , Fatores de Ribosilação do ADP/metabolismo , Brefeldina A/farmacologia , Linhagem Celular Tumoral , Células HEK293 , Humanos , Naftóis/farmacologia , Piridinas/farmacologia , Quinolinas/farmacologia
8.
J Cell Sci ; 130(14): 2251-2265, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28536105

RESUMO

Tether complexes play important roles in endocytic and exocytic trafficking of lipids and proteins. In yeast, the multisubunit transport protein particle (TRAPP) tether regulates endoplasmic reticulum (ER)-to-Golgi and intra-Golgi transport and is also implicated in autophagy. In addition, the TRAPP complex acts as a guanine nucleotide exchange factor (GEF) for Ypt1, which is homologous to human Rab1a and Rab1b. Here, we show that human TRAPPC13 and other TRAPP subunits are critically involved in the survival response to several Golgi-disrupting agents. Loss of TRAPPC13 partially preserves the secretory pathway and viability in response to brefeldin A, in a manner that is dependent on ARF1 and the large GEF GBF1, and concomitant with reduced caspase activation and ER stress marker induction. TRAPPC13 depletion reduces Rab1a and Rab1b activity, impairs autophagy and leads to increased infectivity to the pathogenic bacterium Shigella flexneri in response to brefeldin A. Thus, our results lend support for the existence of a mammalian TRAPPIII complex containing TRAPPC13, which is important for autophagic flux under certain stress conditions.


Assuntos
Antígenos de Neoplasias/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Células A549 , Fator 1 de Ribosilação do ADP/metabolismo , Antibacterianos/farmacologia , Antígenos de Neoplasias/efeitos dos fármacos , Autofagia/fisiologia , Brefeldina A/farmacologia , Disenteria Bacilar/tratamento farmacológico , Disenteria Bacilar/metabolismo , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HEK293 , Células HT29 , Células HeLa , Humanos , Shigella flexneri/efeitos dos fármacos , Proteínas de Transporte Vesicular/antagonistas & inibidores , Proteínas de Transporte Vesicular/efeitos dos fármacos
9.
Cancer Discov ; 3(1): 82-95, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23242809

RESUMO

UNLABELLED: MYC deregulation is common in human cancer. IG-MYC translocations that are modeled in Eµ-Myc mice occur in almost all cases of Burkitt lymphoma as well as in other B-cell lymphoproliferative disorders. Deregulated expression of MYC results in increased mTOR complex 1 (mTORC1) signaling. As tumors with mTORC1 activation are sensitive to mTORC1 inhibition, we used everolimus, a potent and specific mTORC1 inhibitor, to test the requirement for mTORC1 in the initiation and maintenance of Eµ-Myc lymphoma. Everolimus selectively cleared premalignant B cells from the bone marrow and spleen, restored a normal pattern of B-cell differentiation, and strongly protected against lymphoma development. Established Eµ-Myc lymphoma also regressed after everolimus therapy. Therapeutic response correlated with a cellular senescence phenotype and induction of p53 activity. Therefore, mTORC1-dependent evasion of senescence is critical for cellular transformation and tumor maintenance by MYC in B lymphocytes. SIGNIFICANCE: This work provides novel insights into the requirements for MYC-induced oncogenesis by showing that mTORC1 activity is necessary to bypass senescence during transformation of B lymphocytes. Furthermore, tumor eradication through senescence elicited by targeted inhibition of mTORC1 identifies a previously uncharacterized mechanism responsible for significant anticancer activity of rapamycin analogues and serves as proof-of-concept that senescence can be harnessed for therapeutic benefit


Assuntos
Antineoplásicos/uso terapêutico , Linfoma/tratamento farmacológico , Proteínas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/metabolismo , Sirolimo/análogos & derivados , Animais , Linfócitos B/citologia , Linfócitos B/fisiologia , Diferenciação Celular/efeitos dos fármacos , Senescência Celular , Everolimo , Linfoma/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR
10.
Blood ; 114(2): 380-93, 2009 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-19383971

RESUMO

LAQ824 and LBH589 (panobinostat) are histone deacetylase inhibitors (HDACi) developed as cancer therapeutics and we have used the Emu-myc lymphoma model to identify the molecular events required for their antitumor effects. Induction of tumor cell death was necessary for these agents to mediate therapeutic responses in vivo and both HDACi engaged the intrinsic apoptotic cascade that did not require p53. Death receptor pathway blockade had no effect on the therapeutic activities of LAQ824 and LBH589; however, overexpression of Bcl-2 or Bcl-X(L) protected lymphoma cells from HDACi-induced killing and suppressed their therapeutic activities. Deletion of Apaf-1 or Caspase-9 delayed HDACi-induced lymphoma killing in vitro and in vivo, associated with suppression of many biochemical indicators of apoptosis, but did not provide long-term resistance to these agents and failed to inhibit their therapeutic activities. Emu-myc lymphomas lacking a functional apoptosome displayed morphologic and biochemical features of autophagy after treatment with LAQ824 and LBH589, indicating that, in the absence of a complete intrinsic apoptosis pathway involving apoptosome formation, these HDACi can still mediate a therapeutic response. Our data indicate that damage to the mitochondria is the key event necessary for LAQ824 and LBH589 to mediate tumor cell death and a robust therapeutic response.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/farmacologia , Linfoma/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Histona Desacetilases/metabolismo , Humanos , Indóis , Linfoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Transplante de Neoplasias , Panobinostat , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Morte Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Taxa de Sobrevida , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteína Supressora de Tumor p53/metabolismo
11.
Blood ; 113(9): 1982-91, 2009 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-19060243

RESUMO

The apoptotic and therapeutic activities of the histone deacetylase inhibitor (HDACi) vorinostat are blocked by overexpression of Bcl-2 or Bcl-X(L). Herein, we used the small molecule inhibitor ABT-737 to restore sensitivity of Emu-myc lymphomas overexpressing Bcl-2 or Bcl-X(L) to vorinostat and valproic acid (VPA). Combining low-dose ABT-737 with vorinostat or VPA resulted in synergistic apoptosis of these cells. ABT-737 was ineffective against Emu-myc/Mcl-1 and Emu-myc/A1 cells either as a single agent or in combination with HDACi. However, in contrast to the reported binding specificity data, Emu-myc/Bcl-w lymphomas were insensitive to ABT-737 used alone or in combination with HDACi, indicating that the regulatory activity of ABT-737 is restricted to Bcl-2 and Bcl-X(L). Emu-myc lymphomas that expressed Bcl-2 throughout the tumorigenesis process were especially sensitive to ABT-737, while those forced to overexpress Mcl-1 were not. This supports the notion that tumor cells "addicted" to ABT-737 target proteins (ie, Bcl-2 or Bcl-X(L)) are likely to be the most sensitive target cell population. Our studies provide important preclinical data on the binding specificity of ABT-737 and its usefulness against primary hematologic malignancies when used as a single agent and in combination with HDACi.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Compostos de Bifenilo/farmacologia , Inibidores Enzimáticos/administração & dosagem , Inibidores de Histona Desacetilases , Linfoma/tratamento farmacológico , Nitrofenóis/farmacologia , Sulfonamidas/farmacologia , Animais , Compostos de Bifenilo/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Genes bcl-2 , Genes myc , Ácidos Hidroxâmicos/administração & dosagem , Linfoma/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nitrofenóis/administração & dosagem , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Especificidade por Substrato , Sulfonamidas/administração & dosagem , Vorinostat
12.
Proc Natl Acad Sci U S A ; 105(32): 11317-22, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18685088

RESUMO

Histone deacetylase inhibitors (HDACi) and agents such as recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and agonistic anti-TRAIL receptor (TRAIL-R) antibodies are anticancer agents that have shown promise in preclinical settings and in early phase clinical trials as monotherapies. Although HDACi and activators of the TRAIL pathway have different molecular targets and mechanisms of action, they share the ability to induce tumor cell-selective apoptosis. The ability of HDACi to induce expression of TRAIL-R death receptors 4 and 5 (DR4/DR5), and induce tumor cell death via the intrinsic apoptotic pathway provides a molecular rationale to combine these agents with activators of the TRAIL pathway that activate the alternative (death receptor) apoptotic pathway. Herein, we demonstrate that the HDACi vorinostat synergizes with the mouse DR5-specific monoclonal antibody MD5-1 to induce rapid and robust tumor cell apoptosis in vitro and in vivo. Importantly, using a preclinical mouse breast cancer model, we show that the combination of vorinostat and MD5-1 is safe and induces regression of established tumors, whereas single agent treatment had little or no effect. Functional analyses revealed that rather than mediating enhanced tumor cell apoptosis via the simultaneous activation of the intrinsic and extrinsic apoptotic pathways, vorinostat augmented MD5-1-induced apoptosis concomitant with down-regulation of the intracellular apoptosis inhibitor cellular-FLIP (c-FLIP). These data demonstrate that combination therapies involving HDACi and activators of the TRAIL pathway can be efficacious for the treatment of cancer in experimental mouse models.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Inibidores de Histona Desacetilases , Neoplasias Mamárias Experimentais/tratamento farmacológico , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Animais , Anticorpos Monoclonais/farmacologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/biossíntese , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Proteínas de Neoplasias/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Vorinostat
13.
Mol Cancer Ther ; 7(5): 1066-79, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18483296

RESUMO

Histone deacetylase inhibitors (HDACi) are compounds that target the epigenome and cause tumor cell-selective apoptosis. A large number of these agents that have different chemical structures and can target multiple HDACs are being testing in clinical trials and vorinostat is now an approved drug for the treatment of cutaneous T-cell lymphoma. Although these agents are showing promise for the treatment of hematologic malignancies, it is possible that different drugs may have different mechanistic, biological, and therapeutic activities. When comparing an HDACi belonging to the hydroxamic acid class of compounds (vorinostat) with a cyclic tetrapeptide (romidepsin), we showed that these agents regulate the expression of a common set of cellular genes, but certain genes specifically responded to each agent. Using the Emu-myc mouse model of B-cell lymphoma, we showed previously that overexpression of the prosurvival proteins Bcl-2 and Bcl-XL inhibited the apoptotic and therapeutic activities of the vorinostat. Herein, we compared and contrasted the apoptotic-inducing activities of the hydroxamic acid oxamflatin with romidepsin. Like vorinostat, oxamflatin was unable to kill lymphomas overexpressing Bcl-2 and Bcl-XL, indicating that these proteins can generally protect cells against this class of HDACi. In contrast, romidepsin was able to induce apoptosis in lymphomas overexpressing Bcl-2 with delayed kinetics of cell death and could mediate therapeutic responses against these lymphomas. However, romidepsin was inactive when Bcl-XL was overexpressed. These data provide strong support that HDACi of different chemical classes may have subtle yet potentially important differences in their molecular and biological activities.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Apoptose , Depsipeptídeos/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Animais , Antibióticos Antineoplásicos/uso terapêutico , Depsipeptídeos/uso terapêutico , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/uso terapêutico , Ácidos Hidroxâmicos/farmacologia , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo
14.
Cancer Res ; 68(4): 961-4, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18281468

RESUMO

Hedgehog-mediated signaling has been shown to promote growth and dissemination of solid cancers, most prominently basal cell carcinomas and medulloblastoma. Recent findings indicate that hedgehog signals are also important for tumor growth in hematologic malignancies. Hedgehog ligands secreted by stromal cells could elicit Patched/Smoothened-mediated antiapoptotic signaling in mouse B-cell lymphomas. Inhibition of hedgehog signaling induced apoptosis in lymphoma cells and prolonged survival of lymphoma-bearing mice. Depletion of tumor cells proceeded in the absence of p53 via the mitochondrial apoptotic pathway. These and other recently published data on hedgehog inhibition in cancer cells and their implications will be discussed.


Assuntos
Proteínas Hedgehog/metabolismo , Linfoma de Células B/metabolismo , Animais , Humanos , Linfoma de Células B/patologia , Transdução de Sinais , Células Estromais/metabolismo , Células Estromais/patologia
15.
Cell ; 129(2): 423-33, 2007 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-17448999

RESUMO

Bid, a caspase-activated proapoptotic BH3-only protein, is essential for Fas-induced hepatocyte destruction. Recent studies published in Cell produced conflicting results, indicating that loss of Bid either protects or enhances apoptosis induced by DNA damage or replicative stress. To resolve this controversy, we generated novel Bid-deficient mice on an inbred C57BL/6 background and removed the drug-selection cassette from the targeted locus. Nine distinct cell types from these Bid-deficient mice underwent cell-cycle arrest and apoptosis in a manner indistinguishable from control WT cells in response to DNA damage or replicative stress. Moreover, we found that even cells from the original Bid-deficient mice responded normally to these stimuli, indicating that differences in genetic background or the presence of a strong promoter within the targeted locus are unlikely to explain the differences between our results and those reported previously. We conclude that Bid has no role in DNA damage- or replicative stress-induced apoptosis or cell-cycle arrest.


Assuntos
Apoptose , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Ciclo Celular , Dano ao DNA , Animais , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Células Sanguíneas/citologia , Proliferação de Células , Citosol/química , Feminino , Fibroblastos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitógenos/metabolismo
16.
Oncogene ; 24(4): 650-61, 2005 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-15531915

RESUMO

PKCalpha and Ets1 are both associated with breast cancer progression. Our previous studies suggested that these proteins are likely to functionally interact with one another. Here, we show that attenuation of endogenous PKCalpha expression (siPalpha) by RNA interference leads to reduced Ets1 protein expression in a variety of cancer cells. Pulse-chase experiments and treatment with proteasome inhibitor MG-132 revealed that siPalpha interferes with both Ets1 protein synthesis and stability. The effect of siPalpha on Ets1 expression could be partially prevented by KN-93, suggesting that calcium/calmodulin-dependent kinase II (CaMKII), a modulator of Ets1 activity, may play a role in PKCalpha-dependent Ets1 regulation. In contrast, Ets1-regulating kinases ERK1/2 were not found to be involved in this process. To assess the importance of the PKCalpha/Ets1 interaction, we compared the biological responses of MDA-MB-231 cells to PKCalpha- and Ets1-specific siRNAs (siE1). While only siPalpha induced changes in cellular morphology and anchorage-independent growth, both siRNAs similarly affected cellular responses to the antitumor drug mithramycin A and to UV light. Microarray analyses further showed that the expression of a certain set of genes was equally affected by siPalpha and siE1. The data suggest that Ets1 serves as an effector for PKCalpha to fulfil certain functions in cancer cells.


Assuntos
Neoplasias/metabolismo , Plicamicina/análogos & derivados , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Forma Celular , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Plicamicina/farmacologia , Proteína Quinase C/genética , Proteína Quinase C-alfa , Proteína Proto-Oncogênica c-ets-1 , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ets , Interferência de RNA , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Raios Ultravioleta
17.
J Biol Chem ; 280(6): 4476-82, 2005 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-15574417

RESUMO

Granzyme B, a protease released from cytotoxic lymphocytes, has been proposed to induce target cell death by cleaving and activating the pro-apoptotic Bcl-2 family member Bid. It has also been proposed that granzyme B can induce target cell death by activating caspases directly, by cleaving caspase substrates, and/or by cleaving several non-caspase substrates. The relative importance of Bid in granzyme B-induced cell death has therefore remained unclear. Here we report that cells isolated from various tissues of Bid-deficient mice were resistant to granzyme B-induced cell death. Consistent with the proposed role of Bid in regulating mitochondrial outer membrane permeabilization, cytochrome c remained in the mitochondria of Bid-deficient cells treated with granzyme B. Unlike wild type cells, Bid-deficient cells survived and were then able to proliferate normally, demonstrating the critical role for Bid in mediating granzyme B-induced apoptosis.


Assuntos
Apoptose , Proteínas de Transporte/fisiologia , Serina Endopeptidases/metabolismo , Animais , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Morte Celular , Proliferação de Células , Células Cultivadas , Cromo/metabolismo , Citocromos c/metabolismo , Dendritos/metabolismo , Células Dendríticas , Relação Dose-Resposta a Droga , Fibroblastos/metabolismo , Granzimas , Membranas Intracelulares/metabolismo , Linfócitos/metabolismo , Linfoma de Células B/metabolismo , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Fatores de Tempo
18.
Cell Cycle ; 3(6): 779-88, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15153801

RESUMO

Histone deacetylase inhibitors (HDACi) are a promising new class of chemotherapeutic drug currently in early phase clinical trials. A large number of structurally diverse HDACi have been purified or synthesised that mostly inhibit the activity of all eleven class I and II HDACs. While these agents demonstrate many features required for anti-cancer activity such as low toxicity against normal cells and an ability to inhibit tumor cell growth and survival at nanomolar concentrations, their mechanisms of action are largely unknown. Initially, a model was proposed whereby HDACi-mediated transactivation of a specific gene or set of genes was responsible for the inhibition of cell cycle progression or induction of apoptosis. Given that HDACs can regulate the activity of a number of nonhistone proteins and that histone acetylation is important for events such as DNA replication and mitosis that do not directly involve gene transcription, it appears that the initial mechanistic model for HDACi may have been too simple. Herein, we provide an update on the transcription-dependent and -independent events that may be important for the anti-tumor activities of HDACi and discuss the use of these compounds in combination with other chemotherapeutic drugs.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Inibidores de Histona Desacetilases , Neoplasias/tratamento farmacológico , Animais , Humanos
19.
Cell ; 113(6): 717-30, 2003 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-12809603

RESUMO

Efficient engulfment of the intact cell corpse is a critical end point of apoptosis, required to prevent secondary necrosis and inflammation. The presentation of "eat-me" signals on the dying cell is an important part of this process of recognition and engulfment by professional phagocytes. Here, we present evidence that apoptotic cells secrete chemotactic factor(s) that stimulate the attraction of monocytic cells and primary macrophages. The activation of caspase-3 in the apoptotic cell was found to be required for the release of this chemotactic factor(s). The putative chemoattractant was identified as the phospholipid, lysophosphatidylcholine. Further analysis showed that lysophosphatidylcholine was released from apoptotic cells due to the caspase-3 mediated activation of the calcium-independent phospholipase A(2). These data suggest that in addition to eat-me signals, apoptotic cells display attraction signals to ensure the efficient removal of apoptotic cells and prevent postapoptotic necrosis.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Quimiotaxia/fisiologia , Células Eucarióticas/enzimologia , Lisofosfatidilcolinas/metabolismo , Fagócitos/enzimologia , Fagocitose/fisiologia , Animais , Repetição de Anquirina/genética , Células COS , Caspase 3 , Extensões da Superfície Celular/metabolismo , Inibidores Enzimáticos/farmacologia , Células Eucarióticas/metabolismo , Células HT29 , Humanos , Inflamação/enzimologia , Metabolismo dos Lipídeos , Camundongos , Fagócitos/metabolismo , Fosfolipases A/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Transdução de Sinais/fisiologia
20.
Mol Cancer ; 2: 20, 2003 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-12747808

RESUMO

BACKGROUND: Transforming growth factor-beta (TGFbeta) plays an important role in late-stage carcinogenesis by stimulating invasive behavior of cancer cells, promoting neo-angiogenesis and by helping cancer cells to escape surveillance by the immune system. It also supports colonization of the bone by metastatic breast cancer cells by increasing expression of osteolytic parathyroid hormone-related protein (PTHrP). Interfering with TGFbeta signalling may thus weaken the malignant properties of cancer cells. We investigated to what extent two inhibitors, SB-202190 and SB-203580, interfere with TGFbeta-signalling in invasive MDA-MB-231 breast cancer cells. These compounds, formerly used as p38-MAPK-specific inhibitors, were recently also demonstrated to inhibit TGFbeta type I receptor kinase. RESULTS: Our results show that these inhibitors delay the onset of TGFbeta-induced nuclear accumulation of Smad3 and reduces its amplitude. This effect was accompanied by a strong reduction in TGFbeta-responsivess of the slow-responder genes pthrp, pai-1 and upa, while the reactivity of the fast-responder gene smad7 to TGFbeta remained almost unchanged. Neither was the TGFbeta response of the fast-responder ese-1/esx gene, whose expression we found to be strongly downregulated by TGFbeta, affected by the inhibitors. CONCLUSION: The data show that SB-202190 and SB-203580 suppress TGFbeta-dependent activation of genes that are important for the acquisition of invasive behavior, while having no effect on the expression of the natural TGFbeta inhibitor Smad7. This suggests that these compounds are potent inhibitors of malignant behavior of cancer cells.


Assuntos
Núcleo Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Transativadores/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Humanos , Imidazóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína Smad3 , Transativadores/antagonistas & inibidores , Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...