Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Acta Pharmacol Sin ; 40(8): 1067-1075, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30670815

RESUMO

Triple-negative breast cancer (TNBC) is a heterogeneous disease with a poor prognosis due to the lack of an effective targeted therapy. Histone lysine methyltransferases (KMTs) have emerged as attractive drug targets for cancer therapy. However, the function of the majority of KMTs in TNBC has remained largely unknown. In the current study, we found that KMT nuclear receptor binding SET domain protein 2 (NSD2) is overexpressed in TNBC tumors and that its overexpression is associated with poor survival of TNBC patients. NSD2 regulates TNBC cell survival and invasion and is required for tumorigenesis and tumor growth. Mechanistically, NSD2 directly controls the expression of EGFR and ADAM9, a member of the ADAM (a disintegrin and metalloproteinase) family that mediates the release of growth factors, such as HB-EGF. Through its methylase activity, NSD2 overexpression stimulates EGFR-AKT signaling and promotes TNBC cell resistance to the EGFR inhibitor gefitinib. Together, our results identify NSD2 as a major epigenetic regulator in TNBC and provide a rationale for targeting NSD2 alone or in combination with EGFR inhibitors as a targeted therapy for TNBC.


Assuntos
Proteínas ADAM/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/fisiologia , Neoplasias de Mama Triplo Negativas/fisiopatologia , Proteínas ADAM/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Histona-Lisina N-Metiltransferase/genética , Humanos , Proteínas de Membrana/genética , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/fisiopatologia , Proteínas Repressoras/genética , Neoplasias de Mama Triplo Negativas/patologia
2.
Cell Rep Med ; 5(5): 101519, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38692271

RESUMO

Osteosarcoma (OS) is the most common malignant bone tumor with a poor prognosis. Here, we show that the nuclear receptor RORγ may serve as a potential therapeutic target in OS. OS exhibits a hyperactivated oxidative phosphorylation (OXPHOS) program, which fuels the carbon source to promote tumor progression. We found that RORγ is overexpressed in OS tumors and is linked to hyperactivated OXPHOS. RORγ induces the expression of PGC-1ß and physically interacts with it to activate the OXPHOS program by upregulating the expression of respiratory chain component genes. Inhibition of RORγ strongly inhibits OXPHOS activation, downregulates mitochondrial functions, and increases ROS production, which results in OS cell apoptosis and ferroptosis. RORγ inverse agonists strongly suppressed OS tumor growth and progression and sensitized OS tumors to chemotherapy. Taken together, our results indicate that RORγ is a critical regulator of the OXPHOS program in OS and provides an effective therapeutic strategy for this deadly disease.


Assuntos
Neoplasias Ósseas , Mitocôndrias , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Osteossarcoma , Fosforilação Oxidativa , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Osteossarcoma/genética , Humanos , Fosforilação Oxidativa/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Linhagem Celular Tumoral , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/tratamento farmacológico , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Apoptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Ferroptose/genética , Ferroptose/efeitos dos fármacos , Camundongos Nus , Masculino , Proliferação de Células , Proteínas de Ligação a RNA
3.
Prostate ; 73(5): 455-66, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23038103

RESUMO

BACKGROUND: Chromatin regulators ANCCA and EZH2 are overexpressed in prostate cancer and play crucial roles in androgen-stimulated and castration-refractory prostate tumor growth and survival. However, how their expression is regulated in the tumors and whether they play a role in prostate development remains unclear. METHODS: Prostate tissue from different developmental stages of mouse and human were examined by IHC, qRT-PCR and Western for expression of ANCCA, EZH2, and Ki-67. Animals were castrated and T-implanted for the expression response in normal prostate and tumors. siRNA knockdown and ChIP were performed for the mechanism of ANCCA regulation of EZH2. RESULTS: In contrast to their very low level expression in adult prostate, ANCCA and EZH2 are strongly expressed in the epithelium and mesenchyme of mouse and human UGS. Their expression becomes more restricted to epithelial cells during later development and displays a second peak during puberty, which correlates with the proliferative status of the epithelium. Importantly, their expression in normal prostate and tumors is strongly suppressed by castration and markedly induced by testosterone replacement. While androgen suppresses EZH2 in CRPC cells, in LNCaP cells, physiological concentrations of androgen stimulate expression of PRC2 genes (EZH2, SUZ12, and EED), which is mediated by androgen-induced ANCCA and involves E2F and histone H3K4me3 methylase MLL1 complex. CONCLUSION: EZH2 and ANCCA are androgen regulated and strongly expressed in early prostate morphogenesis and during puberty, suggesting their important role in prostate development. Regulation of EZH2 by ANCCA emphasizes bromodomain protein ANCCA as a potential therapeutic target against prostate cancer.


Assuntos
Adenosina Trifosfatases/metabolismo , Androgênios/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexos Multienzimáticos/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Próstata , Neoplasias da Próstata/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Animais , Linhagem Celular Tumoral , Cromatina/enzimologia , Cromatina/genética , Proteínas de Ligação a DNA/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína de Leucina Linfoide-Mieloide/metabolismo , Transplante de Neoplasias , Complexo Repressor Polycomb 2/genética , Gravidez , Próstata/embriologia , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/genética , Maturidade Sexual/fisiologia , Transplante Heterólogo
4.
Biotechnol Prog ; 37(3): e3135, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33527773

RESUMO

The production of recombinant therapeutic proteins from animal or human cell lines entails the risk of endogenous viral contamination from cell substrates and adventitious agents from raw materials and environment. One of the approaches to control such potential viral contamination is to ensure the manufacturing process can adequately clear the potential viral contaminants. Viral clearance for production of human monoclonal antibodies is achieved by dedicated unit operations, such as low pH inactivation, viral filtration, and chromatographic separation. The process development of each viral clearance step for a new antibody production requires significant effort and resources invested in wet laboratory experiments for process characterization studies. Machine learning methods have the potential to help streamline the development and optimization of viral clearance unit operations for new therapeutic antibodies. The current work focuses on evaluating the usefulness of machine learning methods for process understanding and predictive modeling for viral clearance via a case study on low pH viral inactivation.


Assuntos
Anticorpos Monoclonais , Biotecnologia , Aprendizado de Máquina , Inativação de Vírus , Animais , Anticorpos Monoclonais/análise , Anticorpos Monoclonais/isolamento & purificação , Biotecnologia/métodos , Biotecnologia/normas , Células CHO , Cricetinae , Cricetulus , Filtração/métodos , Concentração de Íons de Hidrogênio , Proteínas Recombinantes/análise , Proteínas Recombinantes/isolamento & purificação , Segurança , Vírus/isolamento & purificação
5.
Proc Natl Acad Sci U S A ; 104(46): 18067-72, 2007 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-17998543

RESUMO

AAA+ proteins play crucial roles in diverse biological processes via their ATPase-driven remodeling of macromolecular complexes. Here we report our identification of an evolutionarily conserved AAA+ protein, ANCCA/pro2000, endowed with a bromodomain that is strongly induced by estrogen in human breast cancer cells and is a direct target of protooncogene ACTR/AIB1/SRC-3. We found that ANCCA associates directly with estrogen-bound estrogen receptor (ER) alpha and ACTR. It is selectively recruited, upon estrogen stimulation, to a subset of ERalpha target genes including cyclin D1, c-myc, and E2F1 and is required for their estrogen-induced expression as well as breast cancer cell proliferation. Further studies indicate that ANCCA binds and hydrolyzes ATP and is critical for recruitment of coregulator CBP and histone hyperacetylation at the ER target chromatin. Moreover, mutations at the ATP binding motifs rendered ANCCA defective as a coactivator in mediating estrogen induction of gene expression. Together, our findings reveal an unexpected layer of regulatory mechanism in hormone signaling mediated by ANCCA and suggest that hormone-induced assembly of transcriptional coregulator complexes at chromatin is a process facilitated by AAA+ ATPase proteins.


Assuntos
Adenosina Trifosfatases/metabolismo , Cromatina/metabolismo , Receptor alfa de Estrogênio/metabolismo , Acetilação , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Citometria de Fluxo , Humanos , Hidrólise , RNA Interferente Pequeno
6.
Chem Biol Interact ; 317: 108965, 2020 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-32001260

RESUMO

Endocrine therapies (e.g. tamoxifen and aromatase inhibitors) targeting estrogen action are effective in decreasing mortality of breast cancer. However, their efficacy is limited by intrinsic and acquired resistance. Our previous study demonstrated that overexpression of a histone methyltransferase NSD2 drives tamoxifen resistance in breast cancer cells and that NSD2 is a potential biomarker of tamoxifen resistant breast cancer. Here, we found that DZNep, an indirect inhibitor of histone methyltransferases, potently induces the degradation of NSD2 protein and inhibits the expression of NSD2 target genes (HK2, G6PD, GLUT1 and TIGAR) involved in the pentose phosphate pathway (PPP). DZNep treatment of tamoxifen-resistant breast cancer cells and xenograft tumors also strongly inhibits tumor growth and the cancer cell survival through decreasing cell production of NADPH and glutathione (GSH) and invoking elevated ROS to cause apoptosis. These findings suggest that DZNep-like agents can be developed to target NSD2 histone methyltransferase for effective treatment of tamoxifen-resistant breast cancer.


Assuntos
Adenosina/análogos & derivados , Histona-Lisina N-Metiltransferase/metabolismo , Metiltransferases/antagonistas & inibidores , Proteínas Repressoras/metabolismo , S-Adenosil-Homocisteína/metabolismo , Adenosina/farmacologia , Antígenos Ly , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/genética , Homeostase , Humanos , Oxirredução , Proteólise/efeitos dos fármacos , Espécies Reativas de Oxigênio , Proteínas Repressoras/genética , Tamoxifeno , Ativador de Plasminogênio Tipo Uroquinase
7.
Mol Cell Biol ; 26(10): 3810-23, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16648476

RESUMO

ACTR (also called AIB1 and SRC-3) was identified as a coactivator for nuclear receptors and is linked to multiple types of human cancer due to its frequent overexpression. However, the molecular mechanism of ACTR oncogenicity and its function independent of nuclear receptors remain to be defined. We demonstrate here that ACTR is required for both normal and malignant human cells to effectively enter S phase. RNA interference-mediated depletion and chromatin immunoprecipitation assays show that endogenous ACTR directly controls the expression of genes important for initiation of DNA replication, which include cdc6, cdc25A, MCM7, cyclin E, and Cdk2. Moreover, consistent with its critical role in cell cycle control, ACTR expression appears to be cell cycle regulated, which involves E2F. Interestingly, ACTR is recruited to its own promoter at the G1/S transition and activates its own expression, suggesting a positive feedback mechanism for ACTR action in the control of cell cycle progression and for its aberrant expression in cancers. Importantly, overexpression of ACTR alone transforms human mammary epithelial cells, which requires its association with E2F. These findings reveal a novel role for ACTR in cell cycle control and support the notion that the ability of aberrant ACTR to deregulate the cell cycle through E2F underlies its oncogenicity in human cancers.


Assuntos
Acetiltransferases/metabolismo , Ciclo Celular , Regulação Neoplásica da Expressão Gênica , Regulação da Expressão Gênica , Genes cdc , Proteínas Oncogênicas/metabolismo , Transativadores/metabolismo , Acetiltransferases/genética , Adenoviridae/genética , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Imunoprecipitação da Cromatina , Diploide , Feminino , Fibroblastos/metabolismo , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , Genes Reporter , Glioblastoma/genética , Glioblastoma/patologia , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Histona Acetiltransferases , Humanos , Indóis , Luciferases/metabolismo , Microscopia de Fluorescência , Coativador 3 de Receptor Nuclear , Proteínas Oncogênicas/genética , Regiões Promotoras Genéticas , Proto-Oncogene Mas , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética , Ensaio Tumoral de Célula-Tronco
8.
Nat Commun ; 10(1): 4621, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31604910

RESUMO

Tumor subtype-specific metabolic reprogrammers could serve as targets of therapeutic intervention. Here we show that triple-negative breast cancer (TNBC) exhibits a hyper-activated cholesterol-biosynthesis program that is strongly linked to nuclear receptor RORγ, compared to estrogen receptor-positive breast cancer. Genetic and pharmacological inhibition of RORγ reduces tumor cholesterol content and synthesis rate while preserving host cholesterol homeostasis. We demonstrate that RORγ functions as an essential activator of the entire cholesterol-biosynthesis program, dominating SREBP2 via its binding to cholesterol-biosynthesis genes and its facilitation of the recruitment of SREBP2. RORγ inhibition disrupts its association with SREBP2 and reduces chromatin acetylation at cholesterol-biosynthesis gene loci. RORγ antagonists cause tumor regression in patient-derived xenografts and immune-intact models. Their combination with cholesterol-lowering statins elicits superior anti-tumor synergy selectively in TNBC. Together, our study uncovers a master regulator of the cholesterol-biosynthesis program and an attractive target for TNBC.


Assuntos
Colesterol/biossíntese , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/fisiologia , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Acetilação , Animais , Linhagem Celular Tumoral , Cromatina/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Células MCF-7 , Redes e Vias Metabólicas , Metabolômica , Camundongos , Camundongos Endogâmicos BALB C , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/antagonistas & inibidores
9.
Oncogene ; 38(1): 17-32, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30072740

RESUMO

During the evolution into castration or therapy resistance, prostate cancer cells reprogram the androgen responses to cope with the diminishing level of androgens, and undergo metabolic adaption to the nutritionally deprived and hypoxia conditions. AR (androgen receptor) and PKM2 (pyruvate kinase M2) have key roles in these processes. We report in this study, KDM8/JMJD5, a histone lysine demethylase/dioxygnase, exhibits a novel property as a dual coactivator of AR and PKM2 and as such, it is a potent inducer of castration and therapy resistance. Previously, we showed that KDM8 is involved in the regulation of cell cycle and tumor metabolism in breast cancer cells. Its role in prostate cancer has not been explored. Here, we show that KDM8's oncogenic properties in prostate cancer come from its direct interaction (1) with AR to affect androgen response and (2) with PKM2 to regulate tumor metabolism. The interaction with AR leads to the elevated expression of androgen response genes in androgen-deprived conditions. They include ANCCA/ATAD2 and EZH2, which are directly targeted by KDM8 and involved in sustaining the survival of the cells under hormone-deprived conditions. Notably, in enzalutamide-resistant cells, the expressions of both KDM8 and EZH2 are further elevated, so are neuroendocrine markers. Consequently, EZH2 inhibitors or KDM8 knockdown both resensitize the cells toward enzalutamide. In the cytosol, KDM8 associates with PKM2, the gatekeeper of pyruvate flux and translocates PKM2 into the nucleus, where the KDM8/PKM2 complex serves as a coactivator of HIF-1α to upregulate glycolytic genes. Using shRNA knockdown, we validate KDM8's functions as a regulator for both androgen-responsive and metabolic genes. KDM8 thus presents itself as an ideal therapeutic target for metabolic adaptation and castration-resistance of prostate cancer cells.


Assuntos
Adenocarcinoma/metabolismo , Proteínas de Transporte/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases/fisiologia , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/fisiologia , Neoplasias de Próstata Resistentes à Castração/metabolismo , Receptores Androgênicos/metabolismo , Hormônios Tireóideos/metabolismo , ATPases Associadas a Diversas Atividades Celulares/fisiologia , Transporte Ativo do Núcleo Celular , Adenocarcinoma/patologia , Animais , Benzamidas , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/fisiologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/biossíntese , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Técnicas de Silenciamento de Genes , Glicólise/genética , Xenoenxertos , Histona Desmetilases/biossíntese , Histona Desmetilases/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos Nus , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Nitrilas , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Feniltioidantoína/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/patologia , Mapeamento de Interação de Proteínas , RNA Interferente Pequeno/genética , Receptores Androgênicos/genética , Proteínas de Ligação a Hormônio da Tireoide
10.
Prostate ; 68(16): 1816-26, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18780293

RESUMO

BACKGROUND: Growth of most ablation-resistant prostate cancers (CaPs) is dependent on androgen receptor (AR) activity in chromatin, but cancer cells in these tumors have acquired altered AR activation. It is unclear how the aberrantly activated AR loads onto regulatory regions of AR-targeted genes. The purpose of this study was to assess the AR chromatin loading in an androgen-depleted environment. METHODS: The expression of PSA in androgen-resistant CaP cells was determined using RT-PCR and Western blot analysis. In order to investigate the binding of the AR to the PSA gene regulatory regions, chromatin immunoprecipitation (ChIP) was performed in the androgen-independent cds2 cell line in the presence or absence of androgens. In addition, we examined the involvement of p160 coactivators in the chromatin loading of the AR. RESULTS: It was found that constitutive activation of PSA expression was the result of sustained occupancy by the AR at the regulatory region of this gene. This stable AR loading was not blocked by the AR antagonist bicalutamide. Furthermore, androgen-resistant CaP cells highly expressed both AR and the p160 coactivators and the AR was able to recruit TIF2. Downregulation of TIF2 using short hairpin RNA disrupted the AR loading to the PSA enhancer and subsequently inhibited AR activity. CONCLUSION: Prolonged AR localization to the regulatory regions of AR targeted genes and the recruitment of p160 coactivators are a potential mechanism leading to androgen-independent activation of the AR. Disruption of AR chromatin loading could therefore become an important therapeutic target for this disease.


Assuntos
Adenocarcinoma/metabolismo , Androgênios/metabolismo , Proliferação de Células , Cromatina/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Quinases Associadas a rho/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Anilidas/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cromatina/genética , DNA de Neoplasias/genética , Histona Acetiltransferases/metabolismo , Humanos , Masculino , Nitrilas/farmacologia , Coativador 1 de Receptor Nuclear , Coativador 2 de Receptor Nuclear/genética , Coativador 2 de Receptor Nuclear/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Ligação Proteica , Receptores Androgênicos/genética , Compostos de Tosil/farmacologia , Fatores de Transcrição/metabolismo , Quinases Associadas a rho/genética
11.
Cancer Lett ; 261(1): 64-73, 2008 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-18162290

RESUMO

Overexpression of ACTR/AIB1 is frequently found in different cancers with distant metastasis. To address its possible involvement in tumor metastasis, we performed invasion assays to examine the effect of ACTR alteration on the invasiveness of breast cancer cells (MDA-MB-231 or T-47D) and found that high levels of ACTR are required for their strong invasiveness. Molecular analysis indicates that ACTR functions as a coactivator of AP-1 to up-regulate the expression of matrix metalloproteinases such as MMP-7 and MMP-10 and reduce cell adhesion to specific extracellular matrix proteins. These novel findings provide a mechanistic link between ACTR and MMPs, and suggest that ACTR may also play an important role in cancer progression by facilitating tumor invasion.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Metaloproteinase 10 da Matriz/metabolismo , Metaloproteinase 7 da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Invasividade Neoplásica , Proto-Oncogenes , Fatores de Transcrição/genética , Neoplasias da Mama/patologia , Feminino , Expressão Gênica , Humanos , Coativador 3 de Receptor Nuclear , Proto-Oncogene Mas , Regulação para Cima
12.
Mol Cell Biol ; 24(12): 5157-71, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15169882

RESUMO

Overexpression or amplification of ACTR (also named AIB1, RAC3, p/CIP, TRAM-1, and SRC-3), a member of the p160 family of coactivators for nuclear hormone receptors, has been frequently detected in multiple types of human tumors, including breast cancer. However, its role in cancer cell proliferation and the underlying mechanism are unclear. Here, we show that overexpression of ACTR not only enhances estrogen-stimulated cell proliferation but also, more strikingly, completely negates the cell cycle arrest effect by tamoxifen and pure antiestrogens. Unexpectedly, we found that ACTR directly interacts, through its N-terminal domain, with E2F1 and is recruited to E2F target gene promoters. Elevation of ACTR in quiescent cells strongly stimulates the transcription of a subset of E2F-responsive genes that are associated with the G(1)/S transition. We also demonstrated, by adenovirus vector-mediated RNA interference, that ACTR is required for E2F1-mediated gene expression and the proliferation of estrogen receptor (ER)-negative breast cancer cells. Moreover, the ability of elevated ACTR to promote estrogen-independent cell proliferation depends on the function of E2F1 and the association between ACTR and E2F1, but not ER. Thus, our results reveal an essential role of ACTR in control of breast cancer cell proliferation and implicate the ACTR-E2F1 pathway as a novel mechanism in antiestrogen resistance.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Moduladores de Receptor Estrogênico/farmacologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Sequência de Bases , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , DNA de Neoplasias/genética , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Estradiol/farmacologia , Feminino , Amplificação de Genes , Expressão Gênica , Inativação Gênica , Humanos , Coativador 3 de Receptor Nuclear , RNA Interferente Pequeno/genética , Receptores de Estrogênio/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional
13.
Cancer Lett ; 378(2): 69-79, 2016 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-27164560

RESUMO

Metabolic reprogramming such as the aerobic glycolysis or Warburg effect is well recognized as a common feature of tumorigenesis. However, molecular mechanisms underlying metabolic alterations for tumor therapeutic resistance are poorly understood. Through gene expression profiling analysis we found that histone H3K36 methyltransferase NSD2/MMSET/WHSC1 expression was highly elevated in tamoxifen-resistant breast cancer cell lines and clinical tumors. IHC analysis indicated that NSD2 protein overexpression was associated with the disease recurrence and poor survival. Ectopic expression of NSD2 wild type, but not the methylase-defective mutant, drove endocrine resistance in multiple cell models and xenograft tumors. Mechanistically, NSD2 was recruited to and methylated H3K36me2 at the promoters of key glucose metabolic enzyme genes. Its overexpression coordinately up-regulated hexokinase 2 (HK2) and glucose-6-phosphate dehydrogenase (G6PD), two key enzymes of glycolysis and the pentose phosphate pathway (PPP), as well as TP53-induced glycolysis regulatory phosphatase TIGAR. Consequently, NSD2-driven tamoxifen-resistant cells and tumors displayed heightened PPP activity, elevated NADPH production, and reduced ROS level, without significantly altered glycolysis. These results illustrate a coordinated, epigenetic activation of key glucose metabolic enzymes in therapeutic resistance and nominate methyltransferase NSD2 as a potential therapeutic target for endocrine resistant breast cancer.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Reprogramação Celular , Resistencia a Medicamentos Antineoplásicos , Histona-Lisina N-Metiltransferase/metabolismo , Via de Pentose Fosfato , Proteínas Repressoras/metabolismo , Tamoxifeno/uso terapêutico , Animais , Sítios de Ligação , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Metilação de DNA , Epigênese Genética , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Histona-Lisina N-Metiltransferase/genética , Humanos , Estimativa de Kaplan-Meier , Células MCF-7 , Camundongos Endogâmicos C57BL , Camundongos Nus , Pessoa de Meia-Idade , NADP/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Proteínas Repressoras/genética , Transdução de Sinais , Fatores de Tempo , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cell Death Differ ; 23(11): 1886-1896, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27612013

RESUMO

Recombinant TRAIL and agonistic antibodies to death receptors (DRs) have been in clinical trial but displayed limited anti-cancer efficacy. Lack of functional DR expression in tumors is a major limiting factor. We report here that chromatin regulator KDM4A/JMJD2A, not KDM4B, has a pivotal role in silencing tumor cell expression of both TRAIL and its receptor DR5. In TRAIL-sensitive and -resistant cancer cells of lung, breast and prostate, KDM4A small-molecule inhibitor compound-4 (C-4) or gene silencing strongly induces TRAIL and DR5 expression, and causes TRAIL-dependent apoptotic cell death. KDM4A inhibition also strongly sensitizes cells to TRAIL. C-4 alone potently inhibits tumor growth with marked induction of TRAIL and DR5 expression in the treated tumors and effectively sensitizes them to the newly developed TRAIL-inducer ONC201. Mechanistically, C-4 does not appear to act through the Akt-ERK-FOXO3a pathway. Instead, it switches histone modifying enzyme complexes at promoters of TRAIL and DR5 transcriptional activator CHOP gene by dissociating KDM4A and nuclear receptor corepressor (NCoR)-HDAC complex and inducing the recruitment of histone acetylase CBP. Thus, our results reveal KDM4A as a key epigenetic silencer of TRAIL and DR5 in tumors and establish inhibitors of KDM4A as a novel strategy for effectively sensitizing tumors to TRAIL pathway-based therapeutics.


Assuntos
Antineoplásicos/uso terapêutico , Epigênese Genética , Inativação Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 8/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Epigênese Genética/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Histonas/metabolismo , Humanos , Imidazóis , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Piridinas , Pirimidinas , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo
15.
Nat Med ; 22(5): 488-96, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27019329

RESUMO

The androgen receptor (AR) is overexpressed and hyperactivated in human castration-resistant prostate cancer (CRPC). However, the determinants of AR overexpression in CRPC are poorly defined. Here we show that retinoic acid receptor-related orphan receptor γ (ROR-γ) is overexpressed and amplified in metastatic CRPC tumors, and that ROR-γ drives AR expression in the tumors. ROR-γ recruits nuclear receptor coactivator 1 and 3 (NCOA1 and NCOA3, also known as SRC-1 and SRC-3) to an AR-ROR response element (RORE) to stimulate AR gene transcription. ROR-γ antagonists suppress the expression of both AR and its variant AR-V7 in prostate cancer (PCa) cell lines and tumors. ROR-γ antagonists also markedly diminish genome-wide AR binding, H3K27ac abundance and expression of the AR target gene network. Finally, ROR-γ antagonists suppressed tumor growth in multiple AR-expressing, but not AR-negative, xenograft PCa models, and they effectively sensitized CRPC tumors to enzalutamide, without overt toxicity, in mice. Taken together, these results establish ROR-γ as a key player in CRPC by acting upstream of AR and as a potential therapeutic target for advanced PCa.


Assuntos
Regulação Neoplásica da Expressão Gênica , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Neoplasias de Próstata Resistentes à Castração/genética , Receptores Androgênicos/genética , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzamidas , Sobrevivência Celular/efeitos dos fármacos , Bases de Dados Factuais , Técnicas de Silenciamento de Genes , Glucose-6-Fosfato Isomerase , Humanos , Immunoblotting , Imuno-Histoquímica , Masculino , Camundongos , Transplante de Neoplasias , Nitrilas , Coativador 1 de Receptor Nuclear/metabolismo , Coativador 3 de Receptor Nuclear/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/antagonistas & inibidores , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Piperazinas/farmacologia , Propanóis/farmacologia , Neoplasias de Próstata Resistentes à Castração/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores Androgênicos/metabolismo , Elementos de Resposta , Ensaio Tumoral de Célula-Tronco
16.
PLoS One ; 10(6): e0128229, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26098554

RESUMO

Mammalian cells including human cancer cells are usually transported in cryovials on dry ice or in a liquid nitrogen vapor shipping vessel between different places at long distance. The hazardous nature of dry ice and liquid nitrogen, and the associated high shipping cost strongly limit their routine use. In this study, we tested the viability and properties of cells after being preserved or shipped over long distance in Matrigel mixture for different days. Our results showed that cells mixed with Matrigel at suitable ratios maintained excellent viability (>90%) for one week at room temperature and preserved the properties such as morphology, drug sensitivity and metabolism well, which was comparable to cells cryopreserved in liquid nitrogen. We also sent cells in the Matrigel mixture via FedEx service to different places at ambient temperature. Upon arrival, it was found that over 90% of the cells were viable and grew well after replating. These data collectively suggested that our Matrigel-based method was highly convenient for shipping live cells for long distances in semi-solid gel condition and at ambient temperature.


Assuntos
Colágeno/farmacologia , Laminina/farmacologia , Proteoglicanas/farmacologia , Manejo de Espécimes/métodos , Preservação de Tecido/métodos , Células 3T3 , Animais , Materiais Biocompatíveis/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Combinação de Medicamentos , Géis/farmacologia , Humanos , Células MCF-7 , Camundongos , Temperatura
17.
Mol Cancer Ther ; 14(9): 2090-102, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26141949

RESUMO

Tumor adaptive resistance to therapeutic radiation remains a barrier for further improvement of local cancer control. SIRT3, a member of the sirtuin family of NAD(+)-dependent protein deacetylases in mitochondria, promotes metabolic homeostasis through regulation of mitochondrial protein deacetylation and plays a key role in prevention of cell aging. Here, we demonstrate that SIRT3 expression is induced in an array of radiation-treated human tumor cells and their corresponding xenograft tumors, including colon cancer HCT-116, glioblastoma U87, and breast cancer MDA-MB231 cells. SIRT3 transcriptional activation is due to SIRT3 promoter activation controlled by the stress transcription factor NF-κB. Posttranscriptionally, SIRT3 enzymatic activity is further enhanced via Thr150/Ser159 phosphorylation by cyclin B1-CDK1, which is also induced by radiation and relocated to mitochondria together with SIRT3. Cells expressing Thr150Ala/Ser159Ala-mutant SIRT3 show a reduction in mitochondrial protein lysine deacetylation, Δψm, MnSOD activity, and mitochondrial ATP generation. The clonogenicity of Thr150Ala/Ser159Ala-mutant transfectants is lower and significantly decreased under radiation. Tumors harboring Thr150Ala/Ser159Ala-mutant SIRT3 show inhibited growth and increased sensitivity to in vivo local irradiation. These results demonstrate that enhanced SIRT3 transcription and posttranslational modifications in mitochondria contribute to adaptive radioresistance in tumor cells. CDK1-mediated SIRT3 phosphorylation is a potential effective target to sensitize tumor cells to radiotherapy.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Mitocôndrias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Tolerância a Radiação/genética , Sirtuína 3/genética , Ativação Transcricional , Acetilação , Animais , Proteína Quinase CDC2 , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Mitocôndrias/efeitos da radiação , Proteínas Mitocondriais/metabolismo , Mutação , NF-kappa B/metabolismo , Neoplasias/patologia , Neoplasias/radioterapia , Fosforilação , Sirtuína 3/metabolismo , Transcrição Gênica
18.
Oncotarget ; 6(29): 28440-52, 2015 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-26308378

RESUMO

We have explored the potential for clinical implementation of ATAD2 as a biomarker for aggressive endometrial cancer by investigating to what extent immunohistochemical (IHC) staining for ATAD2 is feasible, reflects clinical phenotype and molecular subgroups of endometrial carcinomas. Increased expression of the ATAD2 gene has been implicated in cancer development and progression in a number of tissues, but few studies have investigated ATAD2 expression using IHC. Here we show that high ATAD2 protein expression is significantly associated with established clinical-pathological variables for aggressive endometrial cancer, also in the subset of estrogen receptor α (ERα) positive tumors. Protein and mRNA expression of ATAD2 were highly correlated (P < 0.001), suggesting that IHC staining may represent a more clinically applicable measure of ATAD2 level in routinely collected formalin fixed paraffin embedded specimens. Gene expression alterations in samples with high ATAD2 expression revealed upregulation of several cancer-related genes (B-MYB, CDCs, E2Fs) and gene sets that previously have been linked to aggressive disease and potential for new targeting therapies. Our results support that IHC staining for ATAD2 may be a clinically applicable biomarker reflecting clinical phenotype and targetable alterations in endometrial carcinomas to be further explored in controlled clinical trials.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Neoplasias do Endométrio/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Transativadores/genética , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/metabolismo , Feminino , Ontologia Genética , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Avaliação de Resultados em Cuidados de Saúde/métodos , Avaliação de Resultados em Cuidados de Saúde/estatística & dados numéricos , Prognóstico , Modelos de Riscos Proporcionais , Estudos Prospectivos , Transativadores/metabolismo
19.
Biomed Res Int ; 2014: 616025, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24967384

RESUMO

Osteosarcoma (OS) is a malignant tumor mainly occurring in children and adolescents. Methotrexate (MTX), a chemotherapy agent, is widely used in treating OS. However, treatment failures are common due to acquired chemoresistance, for which the underlying molecular mechanisms are still unclear. In this study, we report that overexpression of estrogen-related receptor alpha (ERR α ), an orphan nuclear receptor, promoted cell survival and blocked MTX-induced cell death in U2OS cells. We showed that MTX induced ROS production in MTX-sensitive U2OS cells while ERR α effectively blocked the ROS production and ROS associated cell apoptosis. Our further studies demonstrated that ERR α suppressed ROS induction of tumor suppressor P53 and its target genes NOXA and XAF1 which are mediators of P53-dependent apoptosis. In conclusion, this study demonstrated that ERR α plays an important role in the development of MTX resistance through blocking MTX-induced ROS production and attenuating the activation of p53 mediated apoptosis signaling pathway, and points to ERR α as a novel target for improving osteosarcoma therapy.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Metotrexato/farmacologia , Osteossarcoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Estrogênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Adolescente , Proteínas Reguladoras de Apoptose , Neoplasias Ósseas/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Criança , Pré-Escolar , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Osteossarcoma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor ERRalfa Relacionado ao Estrogênio
20.
Mol Cancer Res ; 12(4): 539-49, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24391143

RESUMO

UNLABELLED: Kinesins are a superfamily of motor proteins and often deregulated in different cancers. However, the mechanism of their deregulation has been poorly understood. Through examining kinesin gene family expression in estrogen receptor (ER)-positive breast cancer cells, we found that estrogen stimulation of cancer cell proliferation involves a concerted regulation of specific kinesins. Estrogen strongly induces expression of 19 kinesin genes such as Kif4A/4B, Kif5A/5B, Kif10, Kif11, Kif15, Kif18A/18B, Kif20A/20B, Kif21, Kif23, Kif24, Kif25, and KifC1, whereas suppresses the expression of seven others, including Kif1A, Kif1C, Kif7, and KifC3. Interestingly, the bromodomain protein ANCCA/ATAD2, previously shown to be an estrogen-induced chromatin regulator, plays a crucial role in the up- and downregulation of kinesins by estrogen. Its overexpression drives estrogen-independent upregulation of specific kinesins. Mechanistically, ANCCA (AAA nuclear coregulator cancer associated) mediates E2-dependent recruitment of E2F and MLL1 histone methyltransferase at kinesin gene promoters for gene activation-associated H3K4me3 methylation. Importantly, elevated levels of Kif4A, Kif15, Kif20A, and Kif23 correlate with that of ANCCA in the tumors and with poor relapse-free survival of patients with ER-positive breast cancer. Their knockdown strongly impeded proliferation and induced apoptosis of both tamoxifen-sensitive and resistant cancer cells. Together, the study reveals ANCCA as a key mediator of kinesin family deregulation in breast cancer and the crucial role of multiple kinesins in growth and survival of the tumor cells. IMPLICATIONS: These findings support the development of novel inhibitors of cancer-associated kinesins and their regulator ANCCA for effective treatment of cancers including tamoxifen-resistant breast cancers.


Assuntos
Adenosina Trifosfatases/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Proteínas de Ligação a DNA/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Cinesinas/metabolismo , Tamoxifeno/farmacologia , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Feminino , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/genética , Humanos , Cinesinas/antagonistas & inibidores , Cinesinas/biossíntese , Cinesinas/genética , Células MCF-7 , Transfecção , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA