Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Int J Mol Sci ; 22(8)2021 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-33920782

RESUMO

The role of tumor protein 63 (TP63) in regulating insulin receptor substrate 1 (IRS-1) and other downstream signal proteins in diabetes has not been characterized. RNAs extracted from kidneys of diabetic mice (db/db) were sequenced to identify genes that are involved in kidney complications. RNA sequence analysis showed more than 4- to 6-fold increases in TP63 expression in the diabetic mice's kidneys, compared to wild-type mice at age 10 and 12 months old. In addition, the kidneys from diabetic mice showed significant increases in TP63 mRNA and protein expression compared to WT mice. Mouse proximal tubular cells exposed to high glucose (HG) for 48 h showed significant decreases in IRS-1 expression and increases in TP63, compared to cells grown in normal glucose (NG). When TP63 was downregulated by siRNA, significant increases in IRS-1 and activation of AMP-activated protein kinase (AMPK (p-AMPK-Th172)) occurred under NG and HG conditions. Moreover, activation of AMPK by pretreating the cells with AICAR resulted in significant downregulation of TP63 and increased IRS-1 expression. Ad-cDNA-mediated over-expression of tuberin resulted in significantly decreased TP63 levels and upregulation of IRS-1 expression. Furthermore, TP63 knockdown resulted in increased glucose uptake, whereas IRS-1 knockdown resulted in a decrease in the glucose uptake. Altogether, animal and cell culture data showed a potential role of TP63 as a new candidate gene involved in regulating IRS-1 that may be used as a new therapeutic target to prevent kidney complications in diabetes.


Assuntos
Nefropatias Diabéticas/genética , Transativadores/genética , Regulação para Cima/genética , Adenilato Quinase/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Glicemia/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/sangue , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Túbulos Renais Proximais/patologia , Modelos Biológicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonucleotídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transativadores/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa/farmacologia , Regulação para Cima/efeitos dos fármacos
2.
FASEB J ; 33(1): 373-387, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29975570

RESUMO

Chronic exposure of tubular renal cells to high glucose contributes to tubulointerstitial changes in diabetic nephropathy. In the present study, we identified a new fibrosis gene called galectin-1 (Gal-1), which is highly expressed in tubular cells of kidneys of type 1 and type 2 diabetic mouse models. Gal-1 protein and mRNA expression showed significant increase in kidney cortex of heterozygous Akita+/- and db/db mice compared with wild-type mice. Mouse proximal tubular cells exposed to high glucose showed significant increase in phosphorylation of Akt and Gal-1. We cloned Gal-1 promoter and identified the transcription factor AP4 as binding to the Gal-1 promoter to up-regulate its function. Transfection of cells with plasmid carrying mutations in the binding sites of AP4 to Gal-1 promoter resulted in decreased protein function of Gal-1. In addition, inhibition of Gal-1 by OTX-008 showed significant decrease in p-Akt/AP4 and protein-promoter activity of Gal-1 and fibronectin. Moreover, down-regulation of AP4 by small interfering RNA resulted in a significant decrease in protein expression and promoter activity of Gal-1. We found that kidney of Gal-1-/- mice express very low levels of fibronectin protein. In summary, Gal-1 is highly expressed in kidneys of type 1 and 2 diabetic mice, and AP4 is a major transcription factor that activates Gal-1 under hyperglycemia. Inhibition of Gal-1 by OTX-008 blocks activation of Akt and prevents accumulation of Gal-1, suggesting a novel role of Gal-1 inhibitor as a possible therapeutic target to treat renal fibrosis in diabetes.-Al-Obaidi, N., Mohan, S., Liang, S., Zhao, Z., Nayak, B. K., Li, B., Sriramarao, P., Habib, S. L. Galectin-1 is a new fibrosis protein in type 1 and type 2 diabetes.


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 2/complicações , Fibrose/metabolismo , Galectina 1/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Animais , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Fibronectinas/metabolismo , Fibrose/etiologia , Fibrose/patologia , Glucose/administração & dosagem , Células HEK293 , Humanos , Hipoglicemiantes/administração & dosagem , Insulina/administração & dosagem , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Regiões Promotoras Genéticas
3.
J Biol Chem ; 289(35): 24691-9, 2014 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-25028521

RESUMO

Inactivating mutations of the gene encoding the tricarboxylic acid cycle enzyme fumarate hydratase (FH) have been linked to an aggressive variant of hereditary kidney cancer (hereditary leiomyomatosis and renal cell cancer). These tumors accumulate markedly elevated levels of fumarate. Fumarate is among a growing list of oncometabolites identified in cancers with mutations of genes involved in intermediary metabolism. FH-deficient tumors are notable for their pronounced accumulation of the transcription factor hypoxia inducible factor-1α (HIF-1α) and aggressive behavior. To date, HIF-1α accumulation in hereditary leiomyomatosis and renal cell cancer tumors is thought to result from fumarate-dependent inhibition of prolyl hydroxylases and subsequent evasion from von Hippel-Lindau-dependent degradation. Here, we demonstrate a novel mechanism by which fumarate promotes HIF-1α mRNA and protein accumulation independent of the von Hippel-Lindau pathway. Here we demonstrate that fumarate promotes p65 phosphorylation and p65 accumulation at the HIF-1α promoter through non-canonical signaling via the upstream Tank binding kinase 1 (TBK1). Consistent with these data, inhibition of the TBK1/p65 axis blocks HIF-1α accumulation in cellular models of FH loss and markedly reduces cell invasion of FH-deficient RCC cancer cells. Collectively, our data demonstrate a novel mechanism by which pseudohypoxia is promoted in FH-deficient tumors and identifies TBK1 as a novel putative therapeutic target for the treatment of aggressive fumarate-driven tumors.


Assuntos
Hipóxia Celular , Fumaratos/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Sequência de Bases , Linhagem Celular , Imunoprecipitação da Cromatina , Primers do DNA , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fosforilação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real
4.
Cell Rep ; 43(8): 114527, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39046873

RESUMO

The paracrine actions of adipokine plasminogen activator inhibitor-1 (PAI-1) are implicated in obesity-associated tumorigenesis. Here, we show that PAI-1 mediates extracellular matrix (ECM) signaling via epigenetic repression of DKK1 in endometrial epithelial cells (EECs). While the loss of DKK1 is known to increase ß-catenin accumulation for WNT signaling activation, this epigenetic repression causes ß-catenin release from transmembrane integrins. Furthermore, PAI-1 elicits the disengagement of TIMP2 and SPARC from integrin-ß1 on the cell surface, lifting an integrin-ß1-ECM signaling constraint. The heightened interaction of integrin-ß1 with type 1 collagen (COL1) remodels extracellular fibrillar structures in the ECM. Consequently, the enhanced nanomechanical stiffness of this microenvironment is conducive to EEC motility and neoplastic transformation. The formation of extensively branched COL1 fibrils is also observed in endometrial tumors of patients with obesity. The findings highlight PAI-1 as a contributor to enhanced integrin-COL1 engagement and extensive ECM remodeling during obesity-associated neoplastic development.

5.
Methods Mol Biol ; 2497: 107-115, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35771438

RESUMO

The mitochondrial respiratory chain which carries out the oxidative phosphorylation (OXPHOS) consists of five multi-subunit protein complexes. Emerging evidences suggest that the supercomplexes which further consist of multiple respiratory complexes play important role in regulating OXPHOS function. Dysfunction of the respiratory chain and its regulation has been implicated in various human diseases including neurodegenerative diseases and muscular disorders. Many mouse models have been established which exhibit mitochondrial defects in brain and muscles. Protocols presented here aim to help to analyze the structures of mitochondrial respiratory chain which include the preparation of the tissue samples, isolation of mitochondrial membrane proteins, and analysis of their respiratory complexes by Blue Native Polyacrylamide Gel Electrophoresis (BN-PAGE) in particular.


Assuntos
Membranas Mitocondriais , Fosforilação Oxidativa , Animais , Transporte de Elétrons , Eletroforese em Gel de Poliacrilamida , Camundongos , Eletroforese em Gel de Poliacrilamida Nativa/métodos
6.
Methods Mol Biol ; 2497: 117-128, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35771439

RESUMO

The Seahorse Extracellular Flux Analyzer enables the high-throughput characterization of oxidative phosphorylation capacity based on the electron transport chain organization and regulation with relatively small amount of material. This development over the traditional polarographic Clark-type electrode approaches make it possible to analyze the respiratory features of mitochondria isolated from tissue samples of particular animal models. Here we provide a description of an optimized approach to carry out multi-well measurement of O2 consumption, with the Agilent Seahorse XFe96 analyzer on mouse brain and muscles to determine the tissue-specific oxidative phosphorylation properties. Protocols include the preparation of the tissue samples, isolation of mitochondria, and analysis of their function; in particular, the preparation and optimization of the reagents and samples.


Assuntos
Consumo de Oxigênio , Smegmamorpha , Animais , Transporte de Elétrons , Camundongos , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Polarografia
7.
Cancer Res ; 81(12): 3174-3186, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33863779

RESUMO

Renal cell carcinoma (RCC) mainly originates from renal proximal tubules. Intriguingly, disruption of genes frequently mutated in human RCC samples thus far has only generated RCC originated from other renal tubule parts in mouse models. This hampers our understanding of the pathogenesis of RCC. Here we show that mTOR signaling, often activated in RCC samples, initiates RCC development from renal proximal tubules. Ablation of Tsc1, encoding an mTOR suppressor, in proximal tubule cells led to multiple precancerous renal cysts. mTOR activation increased MEK1 expression and ERK activation, and Mek1 ablation or inhibition diminished cyst formation in Tsc1-deficient mice. mTOR activation also increased MKK6 expression and p38MAPK activation, and ablation of the p38α-encoding gene further enhanced cyst formation and led to RCC with clear cell RCC features. Mechanistically, Tsc1 deletion induced p53 and p16 expression in a p38MAPK-dependent manner, and deleting Tsc1 and Trp53 or Cdkn2a (encoding p16) enhanced renal cell carcinogenesis. Thus, mTOR activation in combination with inactivation of the p38MAPK-p53/p16 pathway drives RCC development from renal proximal tubules. Moreover, this study uncovers previously unidentified mechanisms by which mTOR controls cell proliferation and suggests the MEK-ERK axis to be a potential target for treatment of RCC. SIGNIFICANCE: Mouse modeling studies show that mTOR activation in combination with inactivation of the p38MAPK-p53/p16 axis initiates renal cell carcinoma that mimics human disease, identifying potential therapeutic targets for RCC treatment.


Assuntos
Carcinoma de Células Renais/patologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , MAP Quinase Quinase 1/fisiologia , Proteína Quinase 14 Ativada por Mitógeno/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose , Carcinoma de Células Renais/etiologia , Carcinoma de Células Renais/metabolismo , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Neoplasias Renais/etiologia , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Serina-Treonina Quinases TOR/genética , Células Tumorais Cultivadas
8.
Oncotarget ; 9(68): 32900-32916, 2018 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-30250638

RESUMO

Tuberous sclerosis complex (TSC) disease is associated with tumors in many organs, particularly angiomyolipoma (AML) in the kidneys. Loss or inactivation of TSC1/2 results in high levels of HIF-α activity and VEGF expression. mTOR inhibitor (rapamycin) and the AMPK activator 5-aminoimidazole-4-carboxamide (AICA)-riboside (AICAR) are currently used separately to treat cancer patients. Here, we investigated the effect of a novel combination of rapamycin and AICAR on tumor progression. Our data show that treatment of AML human cells with drug combinations resulted in 5-7-fold increase in cell apoptosis compared to each drug alone. In addition, drug combinations resulted in 4-5-fold decrease in cell proliferation compared to each drug alone. We found that drug combinations abolished Akt and HIF activity in AML cells. The drug combinations resulted in decrease in cell invasion and cell immigration by 70% and 84%, respectively in AML cells. The combined drugs also significantly decreased the VEGF expression compare to each drug alone in AML cells. Drug combinations effectively abolished binding of HIF-2α to the putative Akt site in the nuclear extracts isolated from AML cells. Treatment TSC mice with drug combinations resulted in 75% decrease in tumor number and 88% decrease in tumor volume compared to control TSC mice. This is first evidence that drug combinations are effective in reducing size and number of kidney tumors without any toxic effect on kidney. These data will provide evidence for initiating a new clinical trial for treatment of TSC patients.

9.
Nat Commun ; 8(1): 997, 2017 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-29051480

RESUMO

The molecular mechanisms that couple glycolysis to cancer drug resistance remain unclear. Here we identify an ATP-binding motif within the NADPH oxidase isoform, NOX4, and show that ATP directly binds and negatively regulates NOX4 activity. We find that NOX4 localizes to the inner mitochondria membrane and that subcellular redistribution of ATP levels from the mitochondria act as an allosteric switch to activate NOX4. We provide evidence that NOX4-derived reactive oxygen species (ROS) inhibits P300/CBP-associated factor (PCAF)-dependent acetylation and lysosomal degradation of the pyruvate kinase-M2 isoform (PKM2). Finally, we show that NOX4 silencing, through PKM2, sensitizes cultured and ex vivo freshly isolated human-renal carcinoma cells to drug-induced cell death in xenograft models and ex vivo cultures. These findings highlight yet unidentified insights into the molecular events driving cancer evasive resistance and suggest modulation of ATP levels together with cytotoxic drugs could overcome drug-resistance in glycolytic cancers.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/metabolismo , Resistencia a Medicamentos Antineoplásicos , Metabolismo Energético/fisiologia , Neoplasias Renais/metabolismo , Mitocôndrias/metabolismo , NADPH Oxidase 4/metabolismo , Animais , Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/patologia , Proteínas de Transporte/metabolismo , Células Cultivadas , Metabolismo Energético/efeitos dos fármacos , Etoposídeo/farmacologia , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Hormônios Tireóideos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Fatores de Transcrição de p300-CBP/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
10.
Diabetes ; 65(5): 1387-97, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26908870

RESUMO

Hypoxia-inducible factor (HIF)-1 mediates hypoxia- and chronic kidney disease-induced fibrotic events. Here, we assessed whether HIF-1 blockade attenuates the manifestations of diabetic nephropathy in a type 1 diabetic animal model, OVE26. YC-1 [3-(5'-hydroxymethyl-2'-furyl)-1-benzyl indazole], an HIF-1 inhibitor, reduced whole kidney glomerular hypertrophy, mesangial matrix expansion, extracellular matrix accumulation, and urinary albumin excretion as well as NOX4 protein expression and NADPH-dependent reactive oxygen species production, while blood glucose levels remained unchanged. The role of NOX oxidases in HIF-1-mediated extracellular matrix accumulation was explored in vitro using glomerular mesangial cells. Through a series of genetic silencing and adenoviral overexpression studies, we have defined GLUT1 as a critical downstream target of HIF-1α mediating high glucose-induced matrix expression through the NADPH oxidase isoform, NOX4. Together, our data suggest that pharmacological inhibition of HIF-1 may improve clinical manifestations of diabetic nephropathy.


Assuntos
Diabetes Mellitus Tipo 1/complicações , Nefropatias Diabéticas/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Rim/metabolismo , NADPH Oxidases/metabolismo , Insuficiência Renal/metabolismo , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Linhagem Celular , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/tratamento farmacológico , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/fisiopatologia , Nefropatias Diabéticas/prevenção & controle , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Transportador de Glucose Tipo 1/antagonistas & inibidores , Transportador de Glucose Tipo 1/genética , Hiperglicemia/prevenção & controle , Hipoglicemia/prevenção & controle , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Indazóis/farmacologia , Indazóis/uso terapêutico , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Células Mesangiais/citologia , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/metabolismo , Células Mesangiais/patologia , Camundongos Transgênicos , NADPH Oxidase 4 , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , Estresse Oxidativo/efeitos dos fármacos , Interferência de RNA , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Insuficiência Renal/complicações , Insuficiência Renal/patologia , Insuficiência Renal/prevenção & controle
11.
Bone ; 91: 81-91, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27423464

RESUMO

Multiple myeloma (MM) patients frequently develop tumor-induced bone destruction, yet no therapy completely eliminates the tumor or fully reverses bone loss. Transforming growth factor-ß (TGF-ß) activity often contributes to tumor-induced bone disease, and pre-clinical studies have indicated that TGF-ß inhibition improves bone volume and reduces tumor growth in bone metastatic breast cancer. We hypothesized that inhibition of TGF-ß signaling also reduces tumor growth, increases bone volume, and improves vertebral body strength in MM-bearing mice. We treated myeloma tumor-bearing (immunocompetent KaLwRij and immunocompromised Rag2-/-) mice with a TGF-ß inhibitory (1D11) or control (13C4) antibody, with or without the anti-myeloma drug bortezomib, for 4weeks after inoculation of murine 5TGM1 MM cells. TGF-ß inhibition increased trabecular bone volume, improved trabecular architecture, increased tissue mineral density of the trabeculae as assessed by ex vivo micro-computed tomography, and was associated with significantly greater vertebral body strength in biomechanical compression tests. Serum monoclonal paraprotein titers and spleen weights showed that 1D11 monotherapy did not reduce overall MM tumor burden. Combination therapy with 1D11 and bortezomib increased vertebral body strength, reduced tumor burden, and reduced cortical lesions in the femoral metaphysis, although it did not significantly improve cortical bone strength in three-point bending tests of the mid-shaft femur. Overall, our data provides rationale for evaluating inhibition of TGF-ß signaling in combination with existing anti-myeloma agents as a potential therapeutic strategy to improve outcomes in patients with myeloma bone disease.


Assuntos
Doenças Ósseas/tratamento farmacológico , Doenças Ósseas/etiologia , Osso e Ossos/patologia , Bortezomib/uso terapêutico , Mieloma Múltiplo/complicações , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Doenças Ósseas/patologia , Osso e Ossos/efeitos dos fármacos , Bortezomib/farmacologia , Osso Esponjoso/patologia , Osso Esponjoso/fisiopatologia , Contagem de Células , Linhagem Celular Tumoral , Modelos Animais de Doenças , Quimioterapia Combinada , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/patologia , Osteoblastos/patologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Carga Tumoral/efeitos dos fármacos
12.
Oncogene ; 21(47): 7226-9, 2002 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-12370812

RESUMO

Although it is clear that p53 plays a pivotal role in G1/G2 checkpoints to conserve genomic integrity, its role in S phase checkpoint is less well understood. Recently, it has been reported that p53 is transcriptionally impaired even though it is stabilized during replication blockade. However, the mechanisms underlying this phenomenon are not known. In the present study, it has been shown that p53 accumulates and transactivates its target genes such as p21, gadd45 and bax in response to replication blockade in normal and cancer cells. Lack of transcriptional activation under similar conditions in cells lacking p53 shows that p53-target gene activation during replication blockade is indeed p53-dependent. Further, transactivation of p21 in response to replication blockade by hydroxyurea and aphidicolin is similar to that in response to ionizing radiation except that the latter is more immediate compared to the response to replication blockade. These findings suggest that impairment of transcriptionally active p53 in response to replication blockade is not a general phenomenon.


Assuntos
Replicação do DNA , Genes p53 , Ativação Transcricional , Afidicolina/farmacologia , Células Cultivadas , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos da radiação , Raios gama , Humanos , Hidroxiureia/farmacologia , Transcrição Gênica , Células Tumorais Cultivadas
13.
Oncol Rep ; 9(2): 433-7, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11836622

RESUMO

The frequency of p53 mutations is low and there is evidence of p53 protein overexpression even without p53 mutations in cervical cancers. This suggests that alternative mechanisms other than p53 mutation could be responsible for tumourigenesis of the uterine cervix. Therefore, an attempt has been made in the present investigation to analyze mutation and rearrangement of p53 gene in primary cervical cancers. The results indicated absence of mutation and presence of rearrangement in about 35% of cervical cancer patients. However, p53 overexpression in 50% of patients was demonstrated by immunohistochemistry and Western blot analysis. Further, rearrangement of p53 has been correlated with p53 mRNA and p53 protein status. The results indicated presence of overexpressed p53 protein in the samples with rearranged p53 gene. Thus, it is presumed that rearrangement of p53 might lead to production of defective p53 protein by affecting the level of p53 protein and this might have a role in the process of tumourigenesis. This study reports for the first time rearrangement of p53 in cervical cancers.


Assuntos
Carcinoma de Células Escamosas/genética , Rearranjo Gênico , Genes p53/genética , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/genética , Southern Blotting , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , DNA de Neoplasias/metabolismo , Feminino , Humanos , Técnicas Imunoenzimáticas , Estadiamento de Neoplasias , Mutação Puntual , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia
14.
PLoS One ; 7(1): e30712, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22303451

RESUMO

BACKGROUND: Inflammatory cytokines are detected in the plasma of patients with renal cell carcinoma (RCC) and are associated with poor prognosis. However, the primary cell type involved in producing inflammatory cytokines and the biological significance in RCC remain unknown. Inflammation is associated with oxidative stress, upregulation of hypoxia inducible factor 1-alpha, and production of pro-inflammatory gene products. Solid tumors are often heterogeneous in oxygen tension together suggesting that hypoxia may play a role in inflammatory processes in RCC. Epithelial cells have been implicated in cytokine release, although the stimuli to release and molecular mechanisms by which they are released remain unclear. AMP-activated protein kinase (AMPK) is a highly conserved sensor of cellular energy status and a role for AMPK in the regulation of cell inflammatory processes has recently been demonstrated. METHODS AND PRINCIPAL FINDINGS: We have identified for the first time that interleukin-6 and interleukin-8 (IL-6 and IL-8) are secreted solely from RCC cells exposed to hypoxia. Furthermore, we demonstrate that the NADPH oxidase isoform, Nox4, play a key role in hypoxia-induced IL-6 and IL-8 production in RCC. Finally, we have characterized that enhanced levels of IL-6 and IL-8 result in RCC cell invasion and that activation of AMPK reduces Nox4 expression, IL-6 and IL-8 production, and RCC cell invasion. CONCLUSIONS/SIGNIFICANCE: Together, our data identify novel mechanisms by which AMPK and Nox4 may be linked to inflammation-induced RCC metastasis and that pharmacological activation of AMPK and/or antioxidants targeting Nox4 may represent a relevant therapeutic intervention to reduce IL-6- and IL-8-induced inflammation and cell invasion in RCC.


Assuntos
Carcinoma de Células Renais/enzimologia , Carcinoma de Células Renais/patologia , Interleucina-6/biossíntese , Interleucina-8/biossíntese , Neoplasias Renais/enzimologia , Neoplasias Renais/patologia , NADPH Oxidases/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Antioxidantes/farmacologia , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Humanos , NADPH Oxidase 4 , Invasividade Neoplásica , Ribonucleotídeos/farmacologia
15.
J Biol Chem ; 281(15): 9837-40, 2006 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-16469747

RESUMO

Estrogen receptor-alpha (ERalpha) promotes proliferation of breast cancer cells, whereas tumor suppressor protein p53 impedes proliferation of cells with genomic damage. Whether there is a direct link between these two antagonistic pathways has remained unclear. Here we report that ERalpha binds directly to p53 and represses its function. The activation function-2 (AF-2) domain of ERalpha and the C-terminal regulatory domain of p53 are necessary for the interaction. Knocking down p53 and ERalpha by small interfering RNA elicits opposite effects on p53-target gene expression and cell cycle progression. Remarkably, ionizing radiation that causes genomic damage disrupts the interaction between ERalpha and p53. Ionizing radiation together with ERalpha knock down results in additive effect on transcription of endogenous p53-target gene p21 (CDKN1) in human breast cancer cells. Our findings reveal a novel mechanism for regulating p53 and suggest that suppressing p53 function is an important component in the pro-proliferative role of ERalpha.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Regulação Neoplásica da Expressão Gênica , Proteína Supressora de Tumor p53/metabolismo , Neoplasias da Mama/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Imunoprecipitação da Cromatina , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Receptor alfa de Estrogênio/metabolismo , Genes p53 , Humanos , Immunoblotting , Imunoprecipitação , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Radiação Ionizante , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Ativação Transcricional , Transfecção , Transgenes
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa