Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Exp Cell Res ; 361(2): 201-209, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29056521

RESUMO

Hypertrophic scar (HS) formation is associated with the fibrosis of fibrocytes caused by excessive extracellular matrix (ECM) synthesis and deposition, the initial event of HS formation. Our high throughput screen of miRNA expression profiles identified hsa-miR31-5p, whose transcription level was most differentially in normal skin fibroblasts (NS) and HS among other miRNAs. The level of hsa-miR31-5p in HS was significantly higher than in NS. In-vitro functional experiments showed hsa-miR31-5p knockdown remarkably suppressed the proliferation of hypertrophic scar fibroblasts (HSFBs) under hypoxia, promoted cell invasion, and inhibited the expression of Collagen I and III and Fibronectin (FN), suggesting that hsa-miR31-5p knockdown effectively reduces HS formation caused by excessive ECM synthesis and deposition in HSFBs under hypoxia. Mechanism study showed that the regulation of HS formation by hsa-miR31-5p was mediated by its target gene, factor-inhibiting HIF-1 (FIH): under hypoxia, hsa-miR31-5p down-regulated FIH and thus increased the level of hypoxia inducible factor-1α (HIF-1α), which subsequently activated the HIF-1α fibrosis regulation pathway in HSFBs, and stimulated the proliferation and ECM synthesis in HSFBs, eventually resulting in fibrosis and scar formation. The data also show that knockdown of hsa-miR31-5p in HSFBs impaired the trend of increased proliferation, reduced invasion and excessive ECM synthesis and deposition caused by HIF-1a activation under hypoxia through upregulating FIH, indicating that knockdown of hsa-miR31-5p effectively inhibits the formation of HS. In conclusion, hsa-miR31 -5p plays an important role in HS formation by inhibiting FIH and regulating the HIF-1α pathway. Therefore, hsa-miR31 -5p may be a novel therapeutic target for HS.


Assuntos
Antagomirs/genética , Cicatriz Hipertrófica/genética , Fibroblastos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , MicroRNAs/genética , Oxigenases de Função Mista/genética , Proteínas Repressoras/genética , Antagomirs/metabolismo , Hipóxia Celular , Movimento Celular , Proliferação de Células , Cicatriz Hipertrófica/metabolismo , Cicatriz Hipertrófica/patologia , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibroblastos/patologia , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrose , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Oxigenases de Função Mista/metabolismo , Cultura Primária de Células , Proteínas Repressoras/metabolismo , Transdução de Sinais
2.
Zhonghua Wai Ke Za Zhi ; 56(12): 957-960, 2018 Dec 01.
Artigo em Zh | MEDLINE | ID: mdl-30497123

RESUMO

Acute aortic syndrome(AAS) is a lethal disease with acute onset and a high mortality rate as well as a higher incidence rate especially in an aging population. The diagnostic techniques of AAS have been improving in recent years. Many serum biomarkers have been shown to have the potential of further clinical implication. Advancement of imaging techniques has also improved the accuracy of early diagnosis. Although traditional treatment modality involving open surgery is life-saving, it still has a high mortality rate and a high major morbidity rate. The increasing utilization of endovascular techniques has greatly improved the prognosis of AAS, while it still need further optimization to be applied in different subgroups of patients.


Assuntos
Aneurisma Aórtico , Dissecção Aórtica , Doença Aguda , Dissecção Aórtica/diagnóstico , Dissecção Aórtica/cirurgia , Aneurisma Aórtico/diagnóstico , Aneurisma Aórtico/cirurgia , Doenças da Aorta , Hematoma , Humanos
3.
Ann Oncol ; 28(9): 2135-2141, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28911083

RESUMO

BACKGROUND: Distant metastasis accounts for 90% of deaths from colorectal cancer (CRC). Genomic heterogeneity has been reported in various solid malignancies, but remains largely under-explored in metastatic CRC tumors, especially in primary to metastatic tumor evolution. PATIENTS AND METHODS: We conducted high-depth whole-exome sequencing in multiple regions of matched primary and metastatic CRC tumors. Using a total of 28 tumor, normal, and lymph node tissues, we analyzed inter- and intra-individual heterogeneity, inferred the tumor subclonal architectures, and depicted the subclonal evolutionary routes from primary to metastatic tumors. RESULTS: CRC has significant inter-individual but relatively limited intra-individual heterogeneity. Genomic landscapes were more similar within primary, metastatic, or lymph node tumors than across these types. Metastatic tumors exhibited less intratumor heterogeneity than primary tumors, indicating that single-region sequencing may be adequate to identify important metastasis mutations to guide treatment. Remarkably, all metastatic tumors inherited multiple genetically distinct subclones from primary tumors, supporting a possible polyclonal seeding mechanism for metastasis. Analysis of one patient with the trio samples of primary, metastatic, and lymph node tumors supported a mechanism of synchronous parallel dissemination from the primary to metastatic tumors that was not mediated through lymph nodes. CONCLUSIONS: In CRC, metastatic tumors have different but less heterogeneous genomic landscapes than primary tumors. It is possible that CRC metastasis is, at least partly, mediated through a polyclonal seeding mechanism. These findings demonstrated the rationale and feasibility for identifying and targeting primary tumor-derived metastasis-potent subclones for the prediction, prevention, and treatment of CRC metastasis.


Assuntos
Neoplasias Colorretais/patologia , Sequenciamento do Exoma , Heterogeneidade Genética , Metástase Neoplásica/genética , Neoplasias Colorretais/genética , Humanos , Mutação , Inoculação de Neoplasia
4.
Eur Respir J ; 37(3): 578-86, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20595149

RESUMO

Pulmonary hypertension (PH) and right ventricular (RV) dysfunction associated with heart failure (HF) carry a poor prognosis. Although endothelin receptor antagonists (ERAs) demonstrated benefits in pulmonary arterial hypertension, their efficacy in PH associated with HF was not specifically evaluated. 2 weeks after myocardial infarction (MI) rats received bosentan (100 or 200 mg·kg(-1)·day(-1)) or no treatment for 3 weeks. PH, RV hypertrophy and function as well as lung remodeling and function were evaluated. LV echocardiographic wall motion abnormality and function measured before treatment (2 weeks after MI) and after treatment (5 weeks after MI) were similar in MI control and MI treatment groups. HF induced PH and RV hypertrophy compared with sham: RV systolic pressure 39±5 versus 23±0.8 mmHg and RV/left ventricular+septum weight 52±7 versus 24±0.5% (all p<0.01). Bosentan did not significantly modify these parameters. In addition, bosentan did not improve depressed RV function measured by echocardiograph from the RV myocardial performance index and tricuspid annular plane systolic excursion. The respiratory pressure-volume relationship revealed that HF caused a restrictive lung syndrome with histological lung remodeling and fibrosis, also not improved by bosentan. Dual ERA therapy with bosentan does not reduce PH, RV hypertrophy and lung remodeling and dysfunction associated with ischaemic HF.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Hipertensão Pulmonar/tratamento farmacológico , Sulfonamidas/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Bosentana , Modelos Animais de Doenças , Ecocardiografia/métodos , Antagonistas dos Receptores de Endotelina , Insuficiência Cardíaca/complicações , Ventrículos do Coração/patologia , Hemodinâmica , Hipertensão Pulmonar/complicações , Pulmão/patologia , Pulmão/fisiopatologia , Infarto do Miocárdio/patologia , Prognóstico , Ratos
5.
Mol Cell Biol ; 16(9): 4604-13, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8756616

RESUMO

Expression of vascular endothelial growth factor (VEGF) is induced in cells exposed to hypoxia or ischemia. Neovascularization stimulated by VEGF occurs in several important clinical contexts, including myocardial ischemia, retinal disease, and tumor growth. Hypoxia-inducible factor 1 (HIF-1) is a heterodimeric basic helix-loop-helix protein that activates transcription of the human erythropoietin gene in hypoxic cells. Here we demonstrate the involvement of HIF-1 in the activation of VEGF transcription. VEGF 5'-flanking sequences mediated transcriptional activation of reporter gene expression in hypoxic Hep3B cells. A 47-bp sequence located 985 to 939 bp 5' to the VEGF transcription initiation site mediated hypoxia-inducible reporter gene expression directed by a simian virus 40 promoter element that was otherwise minimally responsive to hypoxia. When reporters containing VEGF sequences, in the context of the native VEGF or heterologous simian virus 40 promoter, were cotransfected with expression vectors encoding HIF-1alpha and HIF-1beta (ARNT [aryl hydrocarbon receptor nuclear translocator]), reporter gene transcription was much greater in both hypoxic and nonhypoxic cells than in cells transfected with the reporter alone. A HIF-1 binding site was demonstrated in the 47-bp hypoxia response element, and a 3-bp substitution eliminated the ability of the element to bind HIF-1 and to activate transcription in response to hypoxia and/or recombinant HIF-1. Cotransfection of cells with an expression vector encoding a dominant negative form of HIF-1alpha inhibited the activation of reporter transcription in hypoxic cells in a dose-dependent manner. VEGF mRNA was not induced by hypoxia in mutant cells that do not express the HIF-1beta (ARNT) subunit. These findings implicate HIF-1 in the activation of VEGF transcription in hypoxic cells.


Assuntos
Hipóxia Celular , Proteínas de Ligação a DNA/fisiologia , Fatores de Crescimento Endotelial/biossíntese , Regulação da Expressão Gênica , Linfocinas/biossíntese , Proteínas Nucleares/fisiologia , Fatores de Transcrição , Transcrição Gênica , Animais , Sequência de Bases , Carcinoma Hepatocelular/patologia , Proteínas de Ligação a DNA/química , Fatores de Crescimento Endotelial/genética , Genes Reporter , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias Hepáticas/patologia , Linfocinas/genética , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/química , Regiões Promotoras Genéticas , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Vírus 40 dos Símios/genética , Transfecção , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
6.
Oncogenesis ; 6(1): e287, 2017 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-28092369

RESUMO

A subset of clear cell renal cell carcinoma (ccRCC) tumors exhibit a HIF1A gene mutation, yielding two ccRCC tumor types, H1H2 type expressing both HIF1α and HIF2α, and H2 type expressing HIF2α, but not functional HIF1α protein. However, it is unclear how the H1H2 type ccRCC tumors escape HIF1's tumor-suppressive activity. The polybromo-1 (PBRM1) gene coding for the BAF180 protein, a component of the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex, is inactivated in 40% ccRCCs, the function and mechanism of BAF180 mutation is unknown. Our previous study indicates that BAF180-containing SWI/SNF chromatin remodeling complex is a co-activator for transcription factor HIF to induce HIF target genes. Thus, our questions are if BAF180 is involved in HIF-mediated hypoxia response and if PBRM1/BAF180 mutation has any association with the HIF1A retention in H1H2 type ccRCC. We report here that BAF180 is mutated in H1H2 ccRCC cell lines and tumors, and BAF180 re-expression in H1H2 ccRCC cell lines reduced cell proliferation/survival, indicating that BAF180 has tumor-suppressive role in these cells. However, BAF180 is expressed in HIF1-deficient H2 ccRCC cell lines and tumors, and BAF180 knockdown in H2 type ccRCC cell lines reduced cell proliferation/survival, indicating that BAF180 has tumor-promoting activity in these cells. In addition, our data show that BAF180 functions as co-activator for HIF1- and HIF2-mediated transcriptional response, and BAF180's tumor-suppressive and -promoting activity in ccRCC cell lines depends on co-expression of HIF1 and HIF2, respectively. Thus, our studies reveal that BAF180 function in ccRCC is context dependent, and that mutation of PBRM1/BAF180 serves as an alternative strategy for ccRCC tumors to reduce HIF1 tumor-suppressive activity in H1H2 ccRCC tumors. Our studies define distinct functional subgroups of ccRCCs based on expression of BAF180, and suggest that BAF180 inhibition may be a novel therapeutic target for patients with H2, but not H1H2, ccRCC tumors.

7.
Oncogene ; 36(18): 2577-2588, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28192397

RESUMO

Epithelial-mesenchymal transition (EMT) has been recognized as a key element of cell migration and invasion in lung cancer; however, the underlying mechanisms are not fully elucidated. Recently, emerging evidence suggest that miRNAs have crucial roles in control of EMT and EMT-associated traits such as migration, invasion and chemoresistance. Here, we found that miR-218 expression levels were significantly downregulated in lung cancer tissues compared with adjacent non-cancerous tissues, and the levels of miR-218 were significantly associated with histological grades and lymph node metastasis. Overexpression of miR-218 inhibited cell migration and invasion as well as the EMT process. Of particular importance, miR-218 was involved in the metastatic process of lung cancer cells in vivo by suppressing local invasion and distant colonization. We identified Slug and ZEB2 as direct functional targets of miR-218. Inverse correlations were observed between miR-218 levels and Slug/ZEB2 levels in cancer tissue samples. In addition, overexpression of miR-218 in H1299 increased chemosensitivity of cells to cisplatin treatment through suppression of Slug and ZEB2. These findings highlight an important role of miR-218 in the regulation of EMT-related traits and metastasis of lung cancer in part by modulation of Slug/ZEB2 signaling, and provide a potential therapeutic strategy by targeting miR-218 in NSCLC.


Assuntos
Proteínas de Homeodomínio/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , Proteínas Repressoras/genética , Fatores de Transcrição da Família Snail/genética , Células A549 , Animais , Movimento Celular/genética , Cisplatino/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Homeobox 2 de Ligação a E-box com Dedos de Zinco
8.
Cancer Res ; 57(23): 5328-35, 1997 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9393757

RESUMO

Adaptation to hypoxia represents an important aspect of tumor progression. Hypoxia-inducible factor 1 (HIF-1) is a transcription factor that mediates essential homeostatic responses to cellular and systemic hypoxia by activating transcription of multiple genes including those encoding glycolytic enzymes and vascular endothelial growth factor (VEGF). In this report, we demonstrate that whereas C-SRC expression is not required for expression of HIF-1 or transcriptional activation of genes encoding VEGF and enolase 1 (ENO1), cells expressing the v-Src oncogene have increased expression of HIF-1, VEGF, and ENO1 under both hypoxic and nonhypoxic conditions. Expression of V-SRC was associated with increased transcription of reporter genes containing cis-acting hypoxia-response elements from the VEGF and ENO1 genes, and this transcriptional activation required an intact HIF-1 binding site. When three rat hepatoma subclones that differed with respect to the level of HIF-1 expression were injected into nude mice, tumor growth correlated with HIF-1 expression, suggesting that HIF-1 may be generally involved in tumor progression. These studies link an oncogene to the induction of HIF-1 expression, thus providing a mechanism for hypoxic adaptation by tumor cells.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Fatores de Crescimento Endotelial/biossíntese , Neoplasias Hepáticas Experimentais/patologia , Linfocinas/biossíntese , Proteínas Nucleares/biossíntese , Proteína Oncogênica pp60(v-src)/biossíntese , Fosfopiruvato Hidratase/biossíntese , Transcrição Gênica , Células 3T3 , Animais , Sequência de Bases , Linhagem Celular , Células Clonais , Progressão da Doença , Embrião de Mamíferos , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias Hepáticas Experimentais/fisiopatologia , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Proteínas Proto-Oncogênicas pp60(c-src)/biossíntese , RNA Mensageiro/biossíntese , Ratos , Proteínas Recombinantes/biossíntese , Fatores de Transcrição/biossíntese , Transfecção , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
9.
Cell Death Dis ; 7: e2216, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-27148689

RESUMO

Kv2.1 as a voltage-gated potassium (Kv) channel subunit has a pivotal role in the regulation of glucose-stimulated insulin secretion (GSIS) and pancreatic ß-cell apoptosis, and is believed to be a promising target for anti-diabetic drug discovery, although the mechanism underlying the Kv2.1-mediated ß-cell apoptosis is obscure. Here, the small molecular compound, ethyl 5-(3-ethoxy-4-methoxyphenyl)-2-(4-hydroxy-3-methoxybenzylidene)-7-methyl-3-oxo-2,3-dihydro-5H-[1,3]thiazolo[3,2-a]pyrimidine-6-carboxylate (SP6616) was discovered to be a new Kv2.1 inhibitor. It was effective in both promoting GSIS and protecting ß cells from apoptosis. Evaluation of SP6616 on either high-fat diet combined with streptozocin-induced type 2 diabetic mice or db/db mice further verified its efficacy in the amelioration of ß-cell dysfunction and glucose homeostasis. SP6616 treatment efficiently increased serum insulin level, restored ß-cell mass, decreased fasting blood glucose and glycated hemoglobin levels, and improved oral glucose tolerance. Mechanism study indicated that the promotion of SP6616 on ß-cell survival was tightly linked to its regulation against both protein kinases C (PKC)/extracellular-regulated protein kinases 1/2 (Erk1/2) and calmodulin(CaM)/phosphatidylinositol 3-kinase(PI3K)/serine/threonine-specific protein kinase (Akt) signaling pathways. To our knowledge, this may be the first report on the underlying pathway responsible for the Kv2.1-mediated ß-cell protection. In addition, our study has also highlighted the potential of SP6616 in the treatment of type 2 diabetes.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Pirimidinas/farmacologia , Canais de Potássio Shab/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Tiazóis/farmacologia , Animais , Células CHO , Calmodulina/antagonistas & inibidores , Calmodulina/genética , Calmodulina/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cricetulus , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Descoberta de Drogas , Regulação da Expressão Gênica , Teste de Tolerância a Glucose , Hipoglicemiantes/química , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinase/genética , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Bloqueadores dos Canais de Potássio/química , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirimidinas/química , Canais de Potássio Shab/genética , Canais de Potássio Shab/metabolismo , Transdução de Sinais , Bibliotecas de Moléculas Pequenas/química , Estreptozocina , Tiazóis/química
10.
Oncogene ; 20(45): 6607-16, 2001 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-11641786

RESUMO

The actin filament-associated protein of 110 kDa (AFAP-110) is a Src binding partner that represents a potential modulator of actin filament integrity in response to cellular signals. Previous reports have demonstrated that AFAP-110 is capable of directly binding and altering actin filaments. Deletion of the leucine zipper motif of AFAP-110 (AFAP-110(Deltalzip)) has been shown to induce a phenotype which resembles Src-transformed cells, by repositioning actin filaments into rosettes. This deletion also mimics a conformational change in AFAP-110 that is detected in Src-transformed cells. The results presented here indicate that unlike AFAP-110, AFAP-110(Deltalzip) is capable of activating cellular tyrosine kinases, including Src family members, and that AFAP-110(Deltalzip) itself is hyperphosphorylated. The newly tyrosine phosphorylated proteins and activated Src-family members appear to be associated with actin-rich lamellipodia. A point mutation that alters the SH3-binding motif of AFAP-110(Deltalzip) prevents it from activating tyrosine kinases and altering actin filament integrity. In addition, a deletion within a pleckstrin homology (PH) domain of AFAP-110(Deltalzip) will also revert its effects upon actin filaments. Lastly, dominant-positive RhoA(V14) will block the ability of AFAP-110(Deltalzip) from inducing actin filament rosettes, but does not inhibit Src activation. Thus, conformational changes in AFAP-110 enable it to activate cellular kinases in a mechanism requiring SH3 and/or PH domain interactions. We hypothesize that cellular signals which alter AFAP-110 conformation, enable it to activate cellular kinases such as cSrc, which then direct changes in actin filament integrity in a Rho-dependent fashion.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Proteínas dos Microfilamentos/fisiologia , Fosfoproteínas/fisiologia , Quinases da Família src/metabolismo , Animais , Células COS , Citoplasma/metabolismo , Citoesqueleto/ultraestrutura , Ativação Enzimática , Zíper de Leucina , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Microscopia de Fluorescência , Modelos Biológicos , Mutação , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilação , Fosfotirosina/metabolismo , Transfecção , Proteína rhoA de Ligação ao GTP/fisiologia , Domínios de Homologia de src
11.
Oncogene ; 34(43): 5482-93, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25703326

RESUMO

Upregulation of the embryonic M2 isoform of pyruvate kinase (PKM2) emerges as a critical player in the cancer development and metabolism, yet the underlying mechanism of PKM2 overexpression remains to be elucidated. Here we demonstrate that IGF-1/IGF-IR regulates PKM2 expression by enhancing HIF-1α-p65 complex binding to PKM2 promoter. PKM2 expression is regulated by miR-148a/152 suppression. PKM2 directly interacts with NF-κB p65 subunit to promote EGR1 expression for regulating miR-148a/152 feedback circuit in normal cells, but not in cancer cells because of the DNA hypermethylation of miR-148a and miR-152 gene promoters. The silencing of miR-148a/152 contributes to the overexpression of PKM2, NF-κB or/and IGF-IR in some cancer cells. We show that disruption of PKM2/NF-κB/miR-148a/152 feedback loop can regulate cancer cell growth and angiogenesis, and is also associated with triple-negative breast cancer (TNBC) phenotype, which may have clinical implication for providing novel biomarker(s) of TNBC and potential therapeutic target(s) in the future.


Assuntos
Proteínas de Transporte/genética , Proteínas de Membrana/genética , MicroRNAs/genética , NF-kappa B/genética , Neovascularização Patológica/genética , Hormônios Tireóideos/genética , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Biomarcadores Tumorais/genética , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Metilação de DNA/genética , Progressão da Doença , Proteína 1 de Resposta de Crescimento Precoce/genética , Células HEK293 , Humanos , Regiões Promotoras Genéticas/genética , Regulação para Cima/genética , Proteínas de Ligação a Hormônio da Tireoide
12.
J Clin Endocrinol Metab ; 86(9): 4284-91, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11549663

RESUMO

We examined GH and GH receptor expression in human leukemic cell lines and leukocytes of normal subjects to elucidate the cell types expressing GH and GH receptor, the individual variations of their expressions, their correlation and the relationships with serum IgG and IGF-I concentrations. In addition, the expression of GH secretagogue receptor, which enhances GH secretion from the anterior pituitary by synthetic GH secretagogues and that of its endogenous ligand, ghrelin, were also examined in these immune cells. GH expression in human leukemic cell lines was observed mainly in B cell lines at both the mRNA and protein level [3.8 +/- 0.2 pg/10(6) cells in Raji and 19.9 +/- 3.3 pg/10(6) cells in Daudi vs. negligible in T cell lines (Jurkat and Hut-78) and in myeloid cell lines (K-562 and HL-60)]. B cells in normal subjects were also found to be the major immune cells expressing GH mRNA, with significant individual variation. GH receptor mRNA expression was detectable in all human leukemic cell lines, although the expression level varied widely among the cell lines and was weaker than that in the liver. On the other hand, GH receptor mRNA expression was mainly found in B cells, with marked individual variation in normal subjects. There was a positive correlation between the mRNA expressions of GH and GH receptor in B cells of normal subjects (r = 0.89; P < 0.001). Single cell RT-PCR revealed that some B cells expressed both GH and GH receptor transcripts, and others expressed only GH. GH/GH receptor expression levels in B cells did not show any correlation with serum IgG and IGF-I levels in normal subjects. Expression of GH secretagogue receptor and ghrelin was detectable in all immune cells regardless of the maturity and cell types with great individual variations. In summary, GH secreted from B cells may act locally on their own receptors, and their variable expressions may be related to individual immune functions. Widespread distribution of ghrelin and GH secretagogue receptor in human immune cells may indicate unknown biological functions other than enhancing GH secretion in the immune system.


Assuntos
Linfócitos B/metabolismo , Hormônio do Crescimento Humano/metabolismo , Neutrófilos/metabolismo , Hormônios Peptídicos , Peptídeos/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G , Receptores da Somatotropina/metabolismo , Linfócitos T/metabolismo , Northern Blotting , Células Cultivadas , Cromatografia em Gel , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática , Grelina , Humanos , Imunoglobulina G/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , RNA Mensageiro/biossíntese , Receptores de Grelina , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Cell Death Dis ; 5: e1407, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25188517

RESUMO

Ovarian cancer is a leading cause of cancer death as diagnosis is frequently delayed to an advanced stage. Effective biomarkers and screening strategies for early detection are urgently needed. In the current study, we identify PSP94 as a key upstream factor in mediating prostasin (a protein previously reported to be overexpressed in ovarian cancer) signaling that regulates prostasin expression and action in ovarian cancer cells. PSP94 is overexpressed in ovarian cancer cell lines and patients, and is significantly correlated with prostasin levels. Signaling pathway analysis demonstrated that both PSP94 and prostasin, as potential upstream regulators of the Lin28b/Let-7 pathway, regulate Lin28b and its downstream partner Let-7 in ovarian cancer cells. Expression of PSP94 and prostasin show a strong correlation with the expression levels of Lin28b/Let-7 in ovarian cancer patients. Thus, PSP94/prostasin axis appears to be linked to the Lin28b/Let-7 loop, a well-known signaling mechanism in oncogenesis in general that is also altered in ovarian cancer. The findings suggest that PSP94 and PSP94/prostasin axis are key factors and potential therapeutic targets or early biomarkers for ovarian cancer.


Assuntos
Neoplasias Ovarianas/patologia , Proteínas Secretadas pela Próstata/metabolismo , Serina Endopeptidases/metabolismo , Biomarcadores Tumorais/sangue , Linhagem Celular Tumoral , Feminino , Humanos , MicroRNAs/metabolismo , Estadiamento de Neoplasias , Neoplasias Ovarianas/metabolismo , Proteínas Secretadas pela Próstata/antagonistas & inibidores , Proteínas Secretadas pela Próstata/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais
14.
Theriogenology ; 80(9): 1088-96, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24055399

RESUMO

Understanding the performances of cloned pigs and their offspring is critical to evaluate the practical applications of somatic cell nuclear transfer. In this study, genetic polymorphism, growth performance, hematological parameters, and reproduction characteristics of cloned Landrace boars were compared with those of controls. In addition, the growth performance of clone offspring was also evaluated. A total of 479 reconstructed embryos were transferred to five recipient pigs and resulted in the delivery of 14 piglets (overall cloning of 2.9%) from two litters. Analyses of microsatellite markers and polymorphisms of the specific genes confirmed that the 14 clones were genetically identical to the nuclear donor and maintained the desirable genotypes. Growth performance of five healthy, phenotypically normal cloned boars from one litter and eight of their male offspring did not differ from age, breed, and management-matched controls. Although some significant differences were observed between cloned and control boars in hematological and serum enzymes, most of these parameters were within the normal range. Cloned boars had less (P < 0.05) normal sperm in the ejaculated boars than in control boars (71.4% vs. 77.9%, respectively), but sperm production (ejaculate volume, sperm concentration, and total sperm) did not differ between these groups. In addition, use of frozen-thawed semen from cloned boars for insemination produced results that seemed comparable to a control. In conclusion, the present study reported that somatic cell nuclear transfer is effective in reproducing preferred genetic traits and has potential applications to conserve elite bloodlines in a routine pig breeding program.


Assuntos
Técnicas de Transferência Nuclear/veterinária , Suínos/genética , Animais , Cruzamento , Clonagem de Organismos/veterinária , Técnicas de Cultura Embrionária/veterinária , Enzimas/sangue , Masculino , Polimorfismo Genético , Reprodução , Análise do Sêmen , Suínos/sangue
15.
Phytomedicine ; 17(8-9): 606-13, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20092987

RESUMO

To elucidate the cytotoxicity mechanism of Ganoderma triterpenes, a chemoproteomic study using five purified ganoderic acids, ganoderic acid F (GAF), ganoderic acid K (GAK), ganoderic B (GAB), ganoderic acid D (GAD) and ganoderic acid AM1 (GAAM1) was conducted. GAF, GAK, GAB, GAD and GAAM1 treatment for 48 h inhibited the proliferation of HeLa human cervical carcinoma cells with IC(50) values of 19.5+/-0.6 microM, 15.1+/-0.5 microM, 20.3+/-0.4 microM, 17.3+/-0.3 microM, 19.8+/-0.7 microM, respectively. The protein expression profiles of HeLa cells treated with each ganoderic acid at dose of 15 microM for 48 h were checked using two-dimensional electrophoresis (2-DE). The possible target-related proteins of ganoderic acids, i.e. proteins with same change tendency in all five ganoderic acids-treated groups compared with control, were identified using MALDI-TOF MS/MS. Twelve proteins including human interleukin-17E, eukaryotic translation initiation factor 5A (eIF5A), peroxiredoxin 2, ubiquilin 2, Cu/Zn-superoxide dismutase, 14-3-3 beta/alpha, TPM4-ALK fusion oncoprotein type 2, PP2A subunit A PR65-alpha isoform, nucleobindin-1, heterogeneous nuclear ribonucleoprotein K, reticulocalbin 1 and chain A of DJ-1 protein were identified. Ganoderic acids might exert their cytotoxicity by altering proteins involved in cell proliferation and/or cell death, carcinogenosis, oxidative stress, calcium signaling and ER stress.


Assuntos
Antineoplásicos/farmacologia , Produtos Biológicos/farmacologia , Carcinoma/metabolismo , Proteoma/efeitos dos fármacos , Reishi/química , Triterpenos/farmacologia , Neoplasias do Colo do Útero/metabolismo , Antineoplásicos/uso terapêutico , Produtos Biológicos/uso terapêutico , Carcinoma/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Eletroforese , Feminino , Células HeLa , Ácidos Heptanoicos/farmacologia , Ácidos Heptanoicos/uso terapêutico , Humanos , Concentração Inibidora 50 , Fitoterapia , Proteínas/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Triterpenos/uso terapêutico , Neoplasias do Colo do Útero/tratamento farmacológico
17.
Biochem Biophys Res Commun ; 212(2): 550-6, 1995 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-7626069

RESUMO

Hypoxia-inducible factor 1 (HIF-1) is a heterodimeric basic helix-loop-helix (bHLH)-PAS DNA-binding protein tightly regulated by cellular oxygen tension. Cellular redox states are related to hypoxia by changes in the expression of redox regulated genes and the generation of reactive oxygen intermediates. Here, we provide evidence that alteration of cellular redox states by treating cells with H2O2 or dithiothreitol impairs hypoxia signaling mechanisms and the expression of HIF-1 alpha protein in hypoxic cells. In addition, HIF-1 DNA-binding activity in vitro is sensitive to oxidizing reagents diamide and H2O2 and the alkylating agent N-ethylmaleimide. The activity of N-ethylmaleimide-inactivated HIF-1 can be partially restored by addition of nuclear extract from nonhypoxic cells.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Expressão Gênica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição , Alquilantes/farmacologia , Hipóxia Celular , Linhagem Celular , Diamida/farmacologia , Ditiotreitol/farmacologia , Etilmaleimida/farmacologia , Peróxido de Hidrogênio/farmacologia , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Oxirredução , Transdução de Sinais/fisiologia
18.
Biochem Biophys Res Commun ; 216(2): 669-75, 1995 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-7488163

RESUMO

Hypoxia-inducible factor 1 (HIF-1) is a heterodimeric bHLH-PAS protein essential for erythropoietin gene transcription in hypoxic cells. Here we show that both 2-aminopurine and sodium fluoride, inhibitors of serine/threonine kinases and phosphatases, respectively, interfered with the hypoxic induction of HIF-1 DNA-binding activity and expression of HIF-1 alpha and HIF-1 beta(ARNT) subunits. Genistein, an inhibitor of tyrosine kinases, completely blocked the synthesis of both HIF-1 subunits as well as HIF-1 DNA-binding activity. Sodium orthovanadate, an inhibitor of tyrosine phosphatases increased the basal level of HIF-1 proteins and HIF-1 activity. These data suggest that protein phosphorylation events play an important role in the hypoxia signal-transduction pathway that leads to synthesis of HIF-1 alpha and HIF-1 beta proteins and the induction of HIF-1 DNA-binding activity.


Assuntos
2-Aminopurina/farmacologia , Proteínas de Ligação a DNA/biossíntese , Inibidores Enzimáticos/farmacologia , Expressão Gênica/efeitos dos fármacos , Proteínas Nucleares/biossíntese , Inibidores de Proteínas Quinases , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Fluoreto de Sódio/farmacologia , Fatores de Transcrição/biossíntese , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/isolamento & purificação , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Immunoblotting , Proteínas Nucleares/isolamento & purificação , Transcrição Gênica/efeitos dos fármacos , Células Tumorais Cultivadas
19.
Proc Natl Acad Sci U S A ; 95(24): 14179-83, 1998 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-9826674

RESUMO

The oncogene p3k, coding for a constitutively active form of phosphatidylinositol 3-kinase (PI 3-kinase; EC 2.7.1.137), strongly enhances myogenic differentiation in cultures of chicken-embryo myoblasts. It increases the size of the myotubes and induces elevated levels of the muscle-specific proteins MyoD, myosin heavy chain, creatine kinase, and desmin. Inhibition of PI 3-kinase activity with LY294002 or with dominant-negative mutants of PI 3-kinase interferes with myogenic differentiation and with the induction of muscle-specific genes. PI 3-kinase is therefore an upstream mediator for the expression of the muscle-specific genes and is both necessary and rate-limiting for the process of myogenesis.


Assuntos
Diferenciação Celular , Músculo Esquelético/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Cromonas/farmacologia , Creatina Quinase/genética , Desmina/genética , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Morfolinas/farmacologia , Músculo Esquelético/enzimologia , Proteína MyoD/genética , Cadeias Pesadas de Miosina/genética , Oncogenes , Fosfatidilinositol 3-Quinases/genética
20.
Proc Natl Acad Sci U S A ; 92(12): 5510-4, 1995 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-7539918

RESUMO

Hypoxia-inducible factor 1 (HIF-1) is found in mammalian cells cultured under reduced O2 tension and is necessary for transcriptional activation mediated by the erythropoietin gene enhancer in hypoxic cells. We show that both HIF-1 subunits are basic-helix-loop-helix proteins containing a PAS domain, defined by its presence in the Drosophila Per and Sim proteins and in the mammalian ARNT and AHR proteins. HIF-1 alpha is most closely related to Sim. HIF-1 beta is a series of ARNT gene products, which can thus heterodimerize with either HIF-1 alpha or AHR. HIF-1 alpha and HIF-1 beta (ARNT) RNA and protein levels were induced in cells exposed to 1% O2 and decayed rapidly upon return of the cells to 20% O2, consistent with the role of HIF-1 as a mediator of transcriptional responses to hypoxia.


Assuntos
Hipóxia Celular , Proteínas de Ligação a DNA/química , Proteínas Nucleares/química , Oxigênio/química , Fatores de Transcrição/química , Sequência de Aminoácidos , Sequência de Bases , DNA Complementar , Proteínas de Ligação a DNA/isolamento & purificação , Sequências Hélice-Alça-Hélice , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Dados de Sequência Molecular , Proteínas Nucleares/isolamento & purificação , RNA/genética , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/isolamento & purificação , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA