Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Pharmacol Res ; 182: 106324, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35750301

RESUMO

The nuclear receptor RORγ is a major driver of autoimmune diseases and certain types of cancer due to its aberrant function in T helper 17 (Th17) cell differentiation and tumor cholesterol metabolism, respectively. Compound screening using the classic receptor-coactivator interaction perturbation scheme led to identification of many small-molecule modulators of RORγ(t). We report here that inverse agonists/antagonists of RORγ such as VTP-43742 derivative VTP-23 and TAK828F, which can potently inhibit the inflammatory gene program in Th17 cells, unexpectedly lack high potency in inhibiting the growth of TNBC tumor cells. In contrast, antagonists such as XY018 and GSK805 that strongly suppress tumor cell growth and survival display only modest activities in reducing Th17-related cytokine expression. Unexpectedly, we found that VTP-23 significantly induces the cholesterol biosynthesis program in TNBC cells. Our further mechanistic analyses revealed that VTP-23 enhances the local chromatin accessibility, H3K27ac mark and the cholesterol master regulator SREBP2 recruitment at the RORγ binding sites, whereas XY018 exerts the opposite activities. Yet, they display similar inhibitory effects on circadian rhythm program. Similar distinctions and contrasting activities between TAK828F and SR2211 in their effects on local chromatin structure at Il17 genes were also observed. Together, our study shows for the first-time that structurally distinct RORγ antagonists possess different or even contrasting activities in tissue/cell-specific manner. Our findings also highlight that the activities at natural chromatin are key determinants of RORγ modulators' tissue selectivity.


Assuntos
Neoplasias de Mama Triplo Negativas , Colesterol/metabolismo , Cromatina/metabolismo , Humanos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Células Th17 , Neoplasias de Mama Triplo Negativas/metabolismo
2.
J Cardiovasc Electrophysiol ; 31(5): 1062-1067, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32108393

RESUMO

INTRODUCTION: The incidence and clinical outcome of pericardial and pleural effusion after cryoballoon ablation (CBA) or radiofrequency catheter ablation (RFCA) of atrial fibrillation (AF) have not been fully investigated. METHODS: A total of 60 patients with paroxysmal AF were treated with either CBA (n = 30) or RFCA (n = 30) groups, with assessment of serum troponin I level, left atrial pulmonary vein computed tomography (CT) angiography and echocardiography within 24 hours before ablation, and serum troponin I level at 12 hours, and chest CT and echocardiography within 24 hours postablation. Repeat chest CT was performed 1 month after the index procedure in patients with pericardial or pleural effusion. RESULTS: With similarly distributed baseline characteristics, the CBA group relative to the RFCA group had postablation: higher serum troponin I level (13.48 vs 1.84 µg/L, P < .001); similarly high pericardial effusion rates on chest CT (80% vs 93.3%, P > .05), with chest CT yielding significantly higher detection rate than echocardiography; similarly high pleural effusion rates on chest CT (73.3% vs 80%, P > .05); and smaller maximum depths on chest CT cross-section of pericardial effusion (5.21 ± 3.37 vs 7.13 ± 2.68 mm, P < .05) and pleural effusion bilaterally (left: 4.16 ± 4.90 vs 6.96 ± 5.42 mm; right: 5.04 ± 4.46 vs 7.55 ± 4.95 mm, both P < .05). The effusions self-resolved within a mean period of 1 month. CONCLUSIONS: Both CBA and RFCA were associated with high rates of pericardial and pleural effusion, with RFCA yielding numerically higher incidence and significantly higher effusion extent, and chest CT significantly higher detection rates than echocardiography.


Assuntos
Fibrilação Atrial/cirurgia , Ablação por Cateter/efeitos adversos , Criocirurgia/efeitos adversos , Derrame Pericárdico/epidemiologia , Derrame Pleural/epidemiologia , Idoso , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/epidemiologia , Biomarcadores/sangue , China/epidemiologia , Ecocardiografia , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Derrame Pericárdico/diagnóstico por imagem , Derrame Pleural/diagnóstico por imagem , Fatores de Risco , Fatores de Tempo , Tomografia Computadorizada por Raios X , Resultado do Tratamento , Troponina I/sangue
3.
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
4.
J Pharmacol Exp Ther ; 365(3): 494-506, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29602831

RESUMO

Noncoding RNAs (ncRNAs) produced in live cells may better reflect intracellular ncRNAs for research and therapy. Attempts were made to produce biologic ncRNAs, but at low yield or success rate. Here we first report a new ncRNA bioengineering technology using more stable ncRNA carrier (nCAR) containing a pre-miR-34a derivative identified by rational design and experimental validation. This approach offered a remarkable higher level expression (40%-80% of total RNAs) of recombinant ncRNAs in bacteria and gave an 80% success rate (33 of 42 ncRNAs). New FPLC and spin-column based methods were also developed for large- and small-scale purification of milligrams and micrograms of recombinant ncRNAs from half liter and milliliters of bacterial culture, respectively. We then used two bioengineered nCAR/miRNAs to demonstrate the selective release of target miRNAs into human cells, which were revealed to be Dicer dependent (miR-34a-5p) or independent (miR-124a-3p), and subsequent changes of miRNome and transcriptome profiles. miRNA enrichment analyses of altered transcriptome confirmed the specificity of nCAR/miRNAs in target gene regulation. Furthermore, nCAR assembled miR-34a-5p and miR-124-3p were active in suppressing human lung carcinoma cell proliferation through modulation of target gene expression (e.g., cMET and CDK6 for miR-34a-5p; STAT3 and ABCC4 for miR-124-3p). In addition, bioengineered miRNA molecules were effective in controlling metastatic lung xenograft progression, as demonstrated by live animal and ex vivo lung tissue bioluminescent imaging as well as histopathological examination. This novel ncRNA bioengineering platform can be easily adapted to produce various ncRNA molecules, and biologic ncRNAs hold the promise as new cancer therapeutics.


Assuntos
Perfilação da Expressão Gênica , Engenharia Genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , MicroRNAs/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica , Neoplasias Pulmonares/patologia , Camundongos
5.
Proc Natl Acad Sci U S A ; 111(52): 18697-702, 2014 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-25512540

RESUMO

Long noncoding RNAs (lncRNAs) have been implicated in a variety of physiological and pathological processes, including cancer. In prostate cancer, prostate cancer gene expression marker 1 (PCGEM1) is an androgen-induced prostate-specific lncRNA whose overexpression is highly associated with prostate tumors. PCGEM1's tumorigenic potential has been recently shown to be in part due to its ability to activate androgen receptor (AR). Here, we report a novel function of PCGEM1 that provides growth advantages for cancer cells by regulating tumor metabolism via c-Myc activation. PCGEM1 promotes glucose uptake for aerobic glycolysis, coupling with the pentose phosphate shunt to facilitate biosynthesis of nucleotide and lipid, and generates NADPH for redox homeostasis. We show that PCGEM1 regulates metabolism at a transcriptional level that affects multiple metabolic pathways, including glucose and glutamine metabolism, the pentose phosphate pathway, nucleotide and fatty acid biosynthesis, and the tricarboxylic acid cycle. The PCGEM1-mediated gene regulation takes place in part through AR activation, but predominantly through c-Myc activation, regardless of hormone or AR status. Significantly, PCGEM1 binds directly to target promoters, physically interacts with c-Myc, promotes chromatin recruitment of c-Myc, and enhances its transactivation activity. We also identified a c-Myc binding domain on PCGEM1 that contributes to the PCGEM1-dependent c-Myc activation and target induction. Together, our data uncover PCGEM1 as a key transcriptional regulator of central metabolic pathways in prostate cancer cells. By being a coactivator for both c-Myc and AR, PCGEM1 reprograms the androgen network and the central metabolism in a tumor-specific way, making it a promising target for therapeutic intervention.


Assuntos
Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Aerobiose/genética , Linhagem Celular Tumoral , Glicólise/genética , Células HEK293 , Humanos , Masculino , NADP/genética , NADP/metabolismo , Via de Pentose Fosfato/genética , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-myc/genética , RNA Longo não Codificante/genética , RNA Neoplásico/genética , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo
6.
J Am Soc Nephrol ; 27(2): 417-27, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26047795

RESUMO

Hypoplastic and/or cystic kidneys have been found in both LDL receptor-related protein 6 (Lrp6)- and ß-catenin-mutant mouse embryos, and these proteins are key molecules for Wnt signaling. However, the underlying mechanisms of Lrp6/ß-catenin signaling in renal development and cystic formation remain poorly understood. In this study, we found evidence that diminished cell proliferation and increased apoptosis occur before cystic dysplasia in the renal primordia of Lrp6-deficient mouse embryos. The expression of Ret proto-oncogene (Ret), a critical receptor for the growth factor glial cell line-derived neurotrophic factor (GDNF), which is required for early nephrogenesis, was dramatically diminished in the mutant renal primordia. The activities of other representative nephrogenic genes, including Lim1, Pax2, Pax8, GDNF, and Wnt11, were subsequently diminished in the mutant renal primordia. Molecular biology experiments demonstrated that Ret is a novel transcriptional target of Wnt/ß-catenin signaling. Wnt agonist lithium promoted Ret expression in vitro and in vivo. Furthermore, Lrp6-knockdown or lithium treatment in vitro led to downregulation or upregulation, respectively, of the phosphorylated mitogen-activated protein kinases 1 and 3, which act downstream of GDNF/Ret signaling. Mice with single and double mutations of Lrp6 and Ret were perinatal lethal and demonstrated gene dosage-dependent effects on the severity of renal hypoplasia during embryogenesis. Taken together, these results suggest that Lrp6-mediated Wnt/ß-catenin signaling modulates or interacts with a signaling network consisting of Ret cascades and related nephrogenic factors for renal development, and the disruption of these genes or signaling activities may cause a spectrum of hypoplastic and cystic kidney disorders.


Assuntos
Rim/crescimento & desenvolvimento , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Rim Displásico Multicístico/etiologia , Proteínas Proto-Oncogênicas c-ret/fisiologia , Transdução de Sinais , Animais , Camundongos , Camundongos Knockout , Rim Displásico Multicístico/genética
7.
Biochim Biophys Acta ; 1853(10 Pt A): 2286-95, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26025680

RESUMO

JMJD5 is a Jumonji C domain-containing demethylase/hydroxylase shown to be essential in embryological development, osteoclastic maturation, circadian rhythm regulation and cancer metabolism. However, its role and underlying mechanisms in oncogenesis remain unclear. Here, we demonstrate that JMJD5 forms complex with the tumor suppressor p53 by interacting with p53 DNA-binding domain (DBD), and negatively regulates its activity. Downregulation of JMJD5 resulted in increased expression of multiple p53 downstream genes, such as the cell cycle inhibitor CDKN1A and DNA repair effector P53R2, only in p53-proficient lung cancer cells. Upon DNA damage, the JMJD5-p53 association decreased, and thereby, promoted p53 recruitment to the target genes and stimulated its transcriptional activity. Furthermore, JMJD5 facilitated the cell cycle progression in a p53-dependent manner under both normal and DNA damage conditions. Depletion of JMJD5 inhibited cell proliferation and enhanced adriamycin-induced cell growth suppression in the presence of p53. Collectively, our results reveal that JMJD5 is a novel binding partner of p53 and it functions as a positive modulator of cell cycle and cell proliferation mainly through the repression of p53 pathway. Our study extends the mechanistic understanding of JMJD5 function in cancer development and implicates JMJD5 as a potential therapeutic target for cancer.


Assuntos
Ciclo Celular , Histona Desmetilases/metabolismo , Neoplasias/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Histona Desmetilases/genética , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Estrutura Terciária de Proteína , Ribonucleotídeo Redutases/genética , Ribonucleotídeo Redutases/metabolismo , Proteína Supressora de Tumor p53/genética
8.
Stem Cells ; 32(1): 45-58, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24115331

RESUMO

The Wnt/ß-catenin pathway is a critical stem cell regulator and plays important roles in neuroepithelial cells during early gestation. However, the role of Wnt/ß-catenin signaling in radial glia, a major neural stem cell population expanded by midgestation, remains poorly understood. This study shows that genetic ablation of ß-catenin with hGFAP-Cre mice inhibits neocortical formation by disrupting radial glial development. Reduced radial glia and intermediate progenitors are found in the ß-catenin-deficient neocortex during late gestation. Increased apoptosis and divergent localization of radial glia in the subventricular zone are also observed in the mutant neocortex. In vivo and in vitro proliferation and neurogenesis as well as oligodendrogenesis by cortical radial glia or by dissociated neural stem cells are significantly defective in the mutants. Neocortical layer patterning is not apparently altered, while astrogliogenesis is ectopically increased in the mutants. At the molecular level, the expression of the transcription factor Pax6 is dramatically diminished in the cortical radial glia and the sphere-forming neural stem cells of ß-catenin-deficient mutants. Chromatin immunoprecipitation and luciferase assays demonstrate that ß-catenin/Tcf complex binds to Pax6 promoter and induces its transcriptional activities. The forced expression of Pax6 through lentiviral transduction partially rescues the defective proliferation and neurogenesis by ß-catenin-deficient neural stem cells. Thus, Pax6 is a novel downstream target of the Wnt/ß-catenin pathway, and ß-catenin/Pax6 signaling plays critical roles in self-renewal and neurogenesis of radial glia/neural stem cells during neocortical development.


Assuntos
Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Neocórtex/citologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , beta Catenina/metabolismo , Animais , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Camundongos , Camundongos Transgênicos , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Fator de Transcrição PAX6 , Transdução de Sinais , Transfecção , Via de Sinalização Wnt , beta Catenina/genética
9.
Am J Pathol ; 183(1): 288-95, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23790802

RESUMO

Prostate cancer (CaP) progresses to a castration-resistant state assisted by multifold molecular changes, most of which involve activation of the androgen receptor (AR). Having previously demonstrated the importance of the Lin28/let-7/Myc axis in CaP, we tested the hypothesis that Lin28 is overexpressed in CaP and that it activates AR and promotes growth of CaP cells. We analyzed human clinical CaP samples for the expression of Lin28 by quantitative real-time RT-PCR, Western blot analysis, and IHC. Growth characteristics of CaP cell lines transiently and stably expressing Lin28 were examined. The clonogenic ability of CaP cells expressing Lin28 was determined by colony formation and soft agar assays. Increase in expression of AR and subsequent increase in transcription of AR-target genes were analyzed by quantitative real-time RT-PCR, luciferase assays, and ELISA. LNCaP cells stably expressing Lin28 were injected into nude mice, and tumorigenesis was monitored. We found that Lin28 is overexpressed in clinical CaP compared to benign prostates. Overexpression of Lin28 enhanced, while down-regulation reduced, growth of CaP cells. Lin28 enhanced the ability of CaP cells to form colonies in anchorage-dependent and anchorage-independent conditions. LNCaP cells stably expressing Lin28 exhibited significantly higher tumorigenic ability in vivo. Lin28 induced expression of the AR and its target genes such as PSA and NKX3.1. Collectively, our findings demonstrate a novel role for Lin28 in CaP development and activation of the AR axis.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores Androgênicos/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Front Immunol ; 15: 1296855, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38449866

RESUMO

Background: Gestational diabetes mellitus (GDM), a transient disease, may lead to short- or long-term adverse influences on maternal and fetal health. Therefore, its potential functions, mechanisms and related molecular biomarkers must be comprehended for the control, diagnosis and treatment of GDM. Methods: The differentially expressed genes (DEGs) were identified using GSE49524 and GSE87295 associated with GDM from the Gene Expression Omnibus database, followed by function enrichment analysis, protein-protein interactions network construction, hub DEGs mining, diagnostic value evaluation and immune infiltration analysis. Finally, hub DEGs, the strongest related to immune infiltration, were screened as immune-related biomarkers. Results: A hundred and seven DEGs were identified between patients with GDM and healthy individuals. Six hub genes with high diagnostic values, including ALDH1A1, BMP4, EFNB2, MME, PLAUR and SLIT2, were identified. Among these, two immune-related genes (PLAUR and SLIT2) with the highest absolute correlation coefficient were considered immune-related biomarkers in GDM. Conclusion: Our study provides a comprehensive analysis of GDM, which would provide a foundation for the development of diagnosis and treatment of GDM.


Assuntos
Diabetes Gestacional , Humanos , Feminino , Gravidez , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/genética , Genes Reguladores , Biomarcadores , Biologia Computacional , Bases de Dados Factuais
11.
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
12.
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
13.
Proc Natl Acad Sci U S A ; 107(21): 9671-6, 2010 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-20457893

RESUMO

Localized chromatin modifications of histone tails play an important role in regulating gene transcription, and aberration of these processes leads to carcinogenesis. Methylated histone lysine residues, a key player in chromatin remodeling, are demethylated by the JmjC class of enzymes. Here we show that JMJD5 (now renamed KDM8), a JmjC family member, demethylates H3K36me2 and is required for cell cycle progression. Chromatin immunoprecipitation assays applied to human genome tiling arrays in conjunction with RNA microarray revealed that KDM8 occupies the coding region of cyclin A1 and directly regulates transcription. Mechanistic analyses showed that KDM8 functioned as a transcriptional activator by inhibiting HDAC recruitment via demethylation of H3K36me2, an epigenetic repressive mark. Tumor array experiments revealed KDM8 is overexpressed in several types of cancer. In addition, loss-of-function studies in MCF7 cells leads to cell cycle arrest. These studies identified KDM8 as an important cell cycle regulator.


Assuntos
Proliferação de Células , Ciclina A1/metabolismo , Histona Desmetilases/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Fases de Leitura Aberta , Acetilação , Linhagem Celular Tumoral , Ciclina A1/genética , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases/genética , Histonas/metabolismo , Humanos , Neoplasias/genética , Interferência de RNA , Transcrição Gênica
14.
Zhonghua Xin Xue Guan Bing Za Zhi ; 41(11): 922-6, 2013 Nov.
Artigo em Zh | MEDLINE | ID: mdl-24370218

RESUMO

OBJECTIVE: To evaluate the clinical characteristics and long-term results of non-pulmonary veins (PV) trigger ablation in patients with paroxysmal atrial fibrillation (AF). METHODS: Eighty-six patients [48 men, mean age (52.3 ± 10.2) years] were included in the study. Circumferential pulmonary vein antrum isolation guided by a 3-D mapping system was performed. Aggressive high right atrium programmed stimulation and burst pacing were made before and after isoproterenol infusion. Additional ablation was performed if other trigger foci were found or other sustained tachycardias could be induced. RESULTS: PV triggers were observed in 59 patients (group I), and non-PV triggers were observed in 27 patients (group II), 12 non-PV triggers were identified during the first procedure. Among them, one was located in the roof of left atrium, 11 were originated from superior vena cava. After a mean follow-up of [37.1 ± 10.4 (range 15-60)] months, the AF recurrence rate was significantly higher in the Group II than in the Group I (55.5% vs. 20.3%, P = 0.001). The number of performed ablation was also significantly in group II than in group I higher difference (1.7 ± 0.8 vs. 1.1 ± 0.4, P < 0.001). In the group II, 15/15 (100%) patients had a repeated ablation procedure for AF recurrence, and 15 patients had new non-PV foci after isoproterenol infusion which were originated from the superior vena cava (n = 11) and coronary sinus (n = 2), respectively. After the second ablation procedure, AF recurrence was observed in three patients, two patients accepted third procedure, the non-PV triggers were located in left atrial septum and coronary sinus, and one patient accepted fourth procedure, the non-PV foci was located in left posterior wall. CONCLUSIONS: Non-PV foci may occur at any age and the main area is located in the super vena cava, Non-PV serves as a major cause of AF recurrence after successful PVAI.


Assuntos
Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/terapia , Ablação por Cateter , Veias Pulmonares/fisiopatologia , Adulto , Idoso , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva , Resultado do Tratamento
15.
Zhonghua Xin Xue Guan Bing Za Zhi ; 41(5): 377-81, 2013 May.
Artigo em Zh | MEDLINE | ID: mdl-24021118

RESUMO

OBJECTIVE: To report the single-center clinical experience of catheter ablation of epicardial accessory pathway associated with coronary sinus musculature. METHODS: The data of 721 cases of left sided accessory pathway ablation were retrospectively analyzed. Ablation in the coronary sinus was performed in 17 (2.4 %) cases [11 males, mean age (37 ± 11) years]. RESULTS: Among the 17 cases, the accessory pathway was successfully ablated in middle cardiac vein and posterior lateral coronary sinus in 11 and 6 cases, respectively. Deverticulum of middle cardiac vein was seen in 2 cases. Mean time required to block the accessory pathway was (4.7 ± 2.7) s. An accessory pathway potential could be recorded at the target site in 10 out of 17 patients (59%). During a mean (21 ± 16) months follow up, only one patient experienced recurrence who was successfully cured by a second ablation session. No procedure related complication was reported. CONCLUSION: About 2.4% of left accessory pathway may have epicardial connection locating at middle cardiac vein or lateral part of the coronary sinus and require epicardial ablation. The epicardial ablation is safe and effective, warrants an excellent long-term results.


Assuntos
Ablação por Cateter , Seio Coronário/cirurgia , Pericárdio/cirurgia , Adulto , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Adulto Jovem
16.
Zhonghua Xin Xue Guan Bing Za Zhi ; 41(3): 233-8, 2013 Mar.
Artigo em Zh | MEDLINE | ID: mdl-23879950

RESUMO

OBJECTIVE: To evaluate the acute and long-term effects of catheter radiofrequency ablation for the treatment of ventricular arrhythmia storm (VAS) post implantable cardioverter-defibrillators (ICD) implantation. METHODS: Acute and long-term effects of catheter radiofrequency ablation for the treatment of VAS post ICD implantation were retrospectively assessed in 11 patients from September 2008 to August 2011. RESULTS: A total of 15 ablation procedures were performed in 11 patients. Six ablation procedures were performed through epicardial approach. In 9 patients, 20 types of ventricular tachycardia (VT) (including 20% hemodynamically unstable VT) were induced during the procedures [mean cycle length (384 ± 141) ms] and polymorphic ventricular tachycardia were induced in 7 patients. The average X-ray fluoroscopy time and procedural time were (26 ± 17) min and (189 ± 60) min, respectively. Complete success, partial success, and failure rates immediately post catheter radiofrequency ablation were 46.7% (7/15), 26.7% (4/15) and 26.7% (4/15), respectively. All patients are alive at follow-up[(2.45 ± 9.6) months after the last catheter ablation] and the complete success, partial success, and failure rates during follow-up were 72.7% (8/11), 9.1% (1/11) and 18.2% (2/11), respectively. CONCLUSION: VAS can be effectively treated by catheter radiofrequency ablation in patients post ICD implantation.


Assuntos
Ablação por Cateter , Desfibriladores Implantáveis/efeitos adversos , Taquicardia Ventricular/cirurgia , Adulto , Idoso , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Taquicardia Ventricular/etiologia , Resultado do Tratamento
17.
Front Pharmacol ; 14: 1146741, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37180705

RESUMO

Natural compounds ursolic acid (UA) and digoxin isolated from fruits and other plants display potent anti-cancer effects in preclinical studies. UA and digoxin have been at clinical trials for treatment of different cancers including prostate cancer, pancreatic cancer and breast cancer. However, they displayed limited benefit to patients. Currently, a poor understanding of their direct targets and mechanisms of action (MOA) severely hinders their further development. We previously identified nuclear receptor RORγ as a novel therapeutic target for castration-resistant prostate cancer (CRPC) and triple-negative breast cancer (TNBC) and demonstrated that tumor cell RORγ directly activates gene programs such as androgen receptor (AR) signaling and cholesterol metabolism. Previous studies also demonstrated that UA and digoxin are potential RORγt antagonists in modulating the functions of immune cells such as Th17 cells. Here we showed that UA displays a strong activity in inhibition of RORγ-dependent transactivation function in cancer cells, while digoxin exhibits no effect at clinically relevant concentrations. In prostate cancer cells, UA downregulates RORγ-stimulated AR expression and AR signaling, whereas digoxin upregulates AR signaling pathway. In TNBC cells, UA but not digoxin alters RORγ-controlled gene programs of cell proliferation, apoptosis and cholesterol biosynthesis. Together, our study reveals for the first-time that UA, but not digoxin, acts as a natural antagonist of RORγ in the cancer cells. Our finding that RORγ is a direct target of UA in cancer cells will help select patients with tumors that likely respond to UA treatment.

18.
Acta Pharm Sin B ; 13(10): 4273-4290, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37799388

RESUMO

During the development of therapeutic microRNAs (miRNAs or miRs), it is essential to define their pharmacological actions. Rather, miRNA research and therapy mainly use miRNA mimics synthesized in vitro. After experimental screening of unique recombinant miRNAs produced in vivo, three lead antiproliferative miRNAs against human NSCLC cells, miR-22-3p, miR-9-5p, and miR-218-5p, were revealed to target folate metabolism by bioinformatic analyses. Recombinant miR-22-3p, miR-9-5p, and miR-218-5p were shown to regulate key folate metabolic enzymes to inhibit folate metabolism and subsequently alter amino acid metabolome in NSCLC A549 and H1975 cells. Isotope tracing studies further confirmed the disruption of one-carbon transfer from serine to folate metabolites by all three miRNAs, inhibition of glucose uptake by miR-22-3p, and reduction of serine biosynthesis from glucose by miR-9-5p and -218-5p in NSCLC cells. With greater activities to interrupt NSCLC cell respiration, glycolysis, and colony formation than miR-9-5p and -218-5p, recombinant miR-22-3p was effective to reduce tumor growth in two NSCLC patient-derived xenograft mouse models without causing any toxicity. These results establish a common antifolate mechanism and differential actions on glucose uptake and metabolism for three lead anticancer miRNAs as well as antitumor efficacy for miR-22-3p nanomedicine, which shall provide insight into developing antimetabolite RNA therapies.

19.
Acta Pharm Sin B ; 13(12): 4934-4944, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38045040

RESUMO

Nuclear transporter importin-ß1 is emerging as an attractive target by virtue of its prevalence in many cancers. However, the lack of druggable inhibitors restricts its therapeutic proof of concept. In the present work, we optimized a natural importin-ß1 inhibitor DD1 to afford an improved analog DD1-Br with better tolerability (>25 folds) and oral bioavailability. DD1-Br inhibited the survival of castration-resistant prostate cancer (CRPC) cells with sub-nanomolar potency and completely prevented tumor growth in resistant CRPC models both in monotherapy (0.5 mg/kg) and in enzalutamide-combination therapy. Mechanistic study revealed that by targeting importin-ß1, DD1-Br markedly inhibited the nuclear accumulation of multiple CRPC drivers, particularly AR-V7, a main contributor to enzalutamide resistance, leading to the integral suppression of downstream oncogenic signaling. This study provides a promising lead for CRPC and demonstrates the potential of overcoming drug resistance in advanced CRPC via targeting importin-ß1.

20.
J Immunother Cancer ; 11(5)2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37147019

RESUMO

BACKGROUND: Emerging data suggest that patients with enzalutamide-treated prostate cancer with increased programmed death-ligand 1 (PD-L1) expression may benefit from anti-PD-L1 treatment. Unfortunately, the Phase III IMbassador250 clinical trial revealed that the combination of atezolizumab (a PD-L1 inhibitor) and enzalutamide failed to extend overall survival in patients with castration-resistant prostate cancer (CRPC). However, the mechanisms underlying treatment failure remain unknown. METHODS: Human CRPC C4-2B cells and murine Myc-CaP cells were chronically exposed to increasing concentrations of enzalutamide and the cells resistant to enzalutamide were referred to as C4-2B MDVR and Myc-CaP MDVR, respectively. The mechanisms of action in drug-resistant prostate cancer cells were determined using RNA sequencing analyses, RNA interference, real-time PCR, western blotting, and co-culturing technologies. Myc-CaP and Myc-CaP MDVR tumors were established in syngeneic FVB mice, and tumor-infiltrating leukocytes were isolated after enzalutamide treatment. The stained immune cells were determined by flow cytometry, and the data were analyzed using FlowJo. RESULTS: Immune-related signaling pathways (interferon alpha/gamma response, inflammatory response, and cell chemotaxis) were suppressed in human enzalutamide-resistant prostate cancer cells. PD-L1 was overexpressed and negatively regulated by androgen receptor signaling in resistant cells and patient with CRPC cohorts. Enzalutamide treatment decreased CD8+ T-cell numbers but increased monocytic myeloid-derived suppressor cell (M-MDSC) populations and PD-L1 expression within murine Myc-CaP tumors. Similarly, chemotaxis and immune response-regulating signaling pathways were suppressed, and PD-L1 expression was also increased using enzalutamide-resistant Myc-CaP MDVR cells. Notably, MDSC populations were significantly increased in Myc-CaP MDVR orthotopic tumors compared with those in Myc-CaP parental tumors. Co-culturing bone marrow cells with Myc-CaP MDVR cells significantly promoted MDSC differentiation and shifted towards M2 macrophage skewing. CONCLUSIONS: Our study suggests that immunosuppressive signaling can be promoted directly by enzalutamide-resistant prostate cancer cells and may be a potential means by which the efficacy of immune checkpoint inhibitors in enzalutamide-resistant prostate cancer is diminished.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Receptores Androgênicos , Animais , Humanos , Masculino , Camundongos , Resistencia a Medicamentos Antineoplásicos , Imunossupressores/uso terapêutico , Feniltioidantoína/farmacologia , Feniltioidantoína/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA