Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Free Radic Biol Med ; 165: 368-384, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33460768

RESUMO

Emerging evidences implicate the contribution of ROS to T cell activation and signaling. The tyrosine kinase, ζ-chain-associated protein of 70 kDa (ZAP70), is essential for T cell development and activation. However, it remains elusive whether a direct redox regulation affects ZAP70 activity upon TCR stimulation. Here, we show that deficiency of non-selenocysteine containing phospholipid hydroperoxide glutathione peroxidase (NPGPx), a redox sensor, results in T cell hyperproliferation and elevated cytokine productions. T cell-specific NPGPx-knockout mice reveal enhanced T-dependent humoral responses and are susceptible to experimental autoimmune encephalomyelitis (EAE). Through proteomic approaches, ZAP70 is identified as the key interacting protein of NPGPx through disulfide bonding. NPGPx is activated by ROS generated from TCR stimulation, and modulates ZAP70 activity through redox switching to reduce ZAP70 recruitment to TCR/CD3 complex in membrane lipid raft, therefore subduing TCR responses. These results reveal a delicate redox mechanism that NPGPx serves as a modulator to curb ZAP70 functions in maintaining T cell homeostasis.


Assuntos
Proteômica , Linfócitos T , Animais , Homeostase , Camundongos , Oxirredução , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo
2.
EMBO Mol Med ; 9(12): 1660-1680, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28993429

RESUMO

Lymph node (LN) metastasis is commonly associated with systemic distant organ metastasis in human breast cancer and is an important prognostic predictor for survival of breast cancer patients. However, whether tumor-draining LNs (TDLNs) play a significant role in modulating the malignancy of cancer cells for distant metastasis remains controversial. Using a syngeneic mouse mammary tumor model, we found that breast tumor cells derived from TDLN have higher malignancy and removal of TDLNs significantly reduced distant metastasis. Up-regulation of oncogenic Il-17rb in cancer cells derived from TDLNs contributes to their malignancy. TGF-ß1 secreted from regulatory T cells (Tregs) in the TDLNs mediated the up-regulation of Il-17rb through downstream Smad2/3/4 signaling. These phenotypes can be abolished by TGF-ß1 neutralization or depletion of Tregs. Consistently, clinical data showed that the up-regulation of IL-17RB in cancer cells from LN metastases correlated with the increased prevalence of Tregs as well as the aggressive growth of tumors in mouse xenograft assay. Together, these results indicate that Tregs in TDLNs play an important role in modulating the malignancy of breast cancer cells for distant metastasis. Blocking IL-17RB expression could therefore be a potential approach to curb the process.


Assuntos
Neoplasias da Mama/patologia , Linfonodos/patologia , Receptores de Interleucina-17/metabolismo , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/radioterapia , Feminino , Humanos , Linfonodos/imunologia , Metástase Linfática , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , RNA Interferente Pequeno/metabolismo , Receptores de Interleucina-17/antagonistas & inibidores , Receptores de Interleucina-17/genética , Transdução de Sinais , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Transplante Homólogo , Células Tumorais Cultivadas , Regulação para Cima
3.
Nat Commun ; 8: 14706, 2017 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-28281525

RESUMO

Adipocytes are the most abundant stromal partners in breast tissue. However, the crosstalk between breast cancer cells and adipocytes has been given less attention compared to cancer-associated fibroblasts. Here we find, through systematic screening, that primary mammary gland-derived adipocytes (MGDAs) promote growth of breast cancer cells that express monocarboxylate transporter 2 (MCT2) both in vitro and in vivo. We show that ß-hydroxybutyrate is secreted by MGDAs and is required to enhance breast cancer cells malignancy in vitro. Consistently, ß-hydroxybutyrate is sufficient to promote tumorigenesis of a mouse xenograft model of MCT2-expressing breast cancer cells. Mechanistically we observe that upon co-culturing with MGDAs or treatment with ß-hydroxybutyrate, breast cancer cells expressing MCT2 increase the global histone H3K9 acetylation and upregulate several tumour-promoting genes. These results suggest that adipocytes promote malignancy of MCT2-expressing breast cancer via ß-hydroxybutyrate potentially by inducing the epigenetic upregulation of tumour-promoting genes.


Assuntos
Ácido 3-Hidroxibutírico/metabolismo , Adipócitos/metabolismo , Neoplasias da Mama/metabolismo , Epigênese Genética , Histonas/genética , Transportadores de Ácidos Monocarboxílicos/genética , Ácido 3-Hidroxibutírico/farmacologia , Acetilação , Adipócitos/patologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Glipicanas/genética , Glipicanas/metabolismo , Histonas/metabolismo , Humanos , Células MCF-7 , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Camundongos , Camundongos Endogâmicos NOD , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transplante de Neoplasias , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Cultura Primária de Células , Pirofosfatases/genética , Pirofosfatases/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
4.
Medicine (Baltimore) ; 95(16): e3407, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27100430

RESUMO

Circulating tumor cells (CTCs) released from a periampullary or pancreatic cancer can be more frequently detected in the portal than the systemic circulation and potentially can be used to identify patients with liver micrometastases. Aims of this study is to determine if CTCs count in portal venous blood of patients with nonmetastatic periampullary or pancreatic adenocarcinoma can be used as a predictor for subsequent liver metastases. CTCs were quantified in portal and peripheral venous blood samples collected simultaneously during pancreaticoduodenectomy in patients with presumed periampullary or pancreatic adenocarcinoma without image-discernible metastasis. Postoperatively patients were monitored for liver metastasis by abdominal magnetic resonance imaging or computed tomography every 3 months for 1 year. Sixty patients with a pathological diagnosis of periampullary or pancreatic adenocarcinoma were included in the study. Multivariate analysis indicated that portal CTC count was a significant predictor for liver metastases within 6 months after surgery. Eleven of 13 patients with a high portal CTCs count (defined as >112 CMx Platform estimated CTCs in 2 mL blood) developed liver metastases within 6 months after surgery. In contrast, only 6 of 47 patients with a low portal CTC count developed liver metastases (P < 0.0001). A value of 112 CMx Platform estimated CTCs had 64.7% sensitivity and 95.4% specificity to predict liver metastases within 6 months after surgery. We concluded that a high CTC count in portal venous blood collected during pancreaticoduodenectomy in patients with periampullary or pancreatic adenocarcinoma without metastases detected by currently available imaging tools is a significant predictor for liver metastases within 6 months after surgery.


Assuntos
Adenocarcinoma/secundário , Neoplasias Hepáticas/secundário , Estadiamento de Neoplasias/métodos , Células Neoplásicas Circulantes/patologia , Neoplasias Pancreáticas/patologia , Adenocarcinoma/diagnóstico , Adenocarcinoma/cirurgia , Idoso , Biópsia , Contagem de Células , Feminino , Seguimentos , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/cirurgia , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/cirurgia , Pancreaticoduodenectomia , Veia Porta , Valor Preditivo dos Testes , Estudos Prospectivos
5.
Nucleic Acids Res ; 43(19): 9393-404, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26446990

RESUMO

Non-selenocysteine-containing phospholipid hydroperoxide glutathione peroxidase (NPGPx or GPx7) is an oxidative stress sensor that modulates the antioxidative activity of its target proteins through intermolecular disulfide bond formation. Given NPGPx's role in protecting cells from oxidative damage, identification of the oxidative stress-induced protein complexes, which forms with key stress factors, may offer novel insight into intracellular reactive oxygen species homeostasis. Here, we show that NPGPx forms a disulfide bond with the translational regulator cytoplasmic polyadenylation element-binding protein 2 (CPEB2) that results in negative regulation of hypoxia-inducible factor 1-alpha (HIF-1α) RNA translation. In NPGPx-proficient cells, high oxidative stress that disrupts this bonding compromises the association of CPEB2 with HIF-1α RNA, leading to elevated HIF-1α RNA translation. NPGPx-deficient cells, in contrast, demonstrate increased HIF-1α RNA translation under normoxia with both impaired induction of HIF-1α synthesis and blunted HIF-1α-programmed transcription following oxidative stress. Together, these results reveal a molecular mechanism for how NPGPx mediates CPEB2-controlled HIF-1α RNA translation in a redox-sensitive manner.


Assuntos
Proteínas de Transporte/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Estresse Oxidativo , Biossíntese de Proteínas , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas de Transporte/genética , Células Cultivadas , Cisteína/análise , Dissulfetos/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA/química , Ratos , Transcrição Gênica
6.
Oncotarget ; 6(34): 36278-91, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26462147

RESUMO

G-protein-coupled receptor kinase interacting protein 1 (GIT1) is participated in cell movement activation, which is a fundamental process during tissue development and cancer progression. GIT1/PIX forming a functional protein complex that contributes to Rac1/Cdc42 activation, resulting in increasing cell mobility. Although the importance of Rac1/Cdc42 activation is well documented in cancer aggressiveness, the clinical importance of GIT1 remains largely unknown. Here, we investigated the clinical significance of GIT1 expression in non-small-cell lung cancer (NSCLC) and also verified the importance of GIT1-Rac1/Cdc42 axis in stimulating NSCLC cell mobility. The result indicated higher GIT1 expression patients had significantly poorer prognoses in disease-free survival (DFS) and overall survival (OS) compared with lower GIT1 expression patients. Higher GIT1 expression was an independent prognostic factor by multivariate analysis and associated with migration/invasion of NSCLC cells in transwell assay. In vivo studies indicated that GIT1 promotes metastasis of NSCLC cells. Finally, GIT1 was found to stimulate migration/invasion by altering the activity of Rac1/Cdc42 in NSCLC cells. Together, the GIT1 expression is associated with poor prognosis in patients with NSCLC. GIT1 is critical for the invasiveness of NSCLC cells through stimulating the activity of Rac1/Cdc42.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neoplasias Pulmonares/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Idoso , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Prognóstico , Transdução de Sinais , Transfecção
7.
J Exp Med ; 212(3): 333-49, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25732306

RESUMO

Pancreatic cancer has an extremely high mortality rate due to its aggressive metastatic nature. Resolving the underlying mechanisms will be crucial for treatment. Here, we found that overexpression of IL-17B receptor (IL-17RB) strongly correlated with postoperative metastasis and inversely correlated with progression-free survival in pancreatic cancer patients. Consistently, results from ex vivo experiments further validated that IL-17RB and its ligand, IL-17B, plays an essential role in pancreatic cancer metastasis and malignancy. Signals from IL-17B-IL-17RB activated CCL20/CXCL1/IL-8/TFF1 chemokine expressions via the ERK1/2 pathway to promote cancer cell invasion, macrophage and endothelial cell recruitment at primary sites, and cancer cell survival at distant organs. Treatment with a newly derived monoclonal antibody against IL-17RB blocked tumor metastasis and promoted survival in a mouse xenograft model. These findings not only illustrate a key mechanism underlying the highly aggressive characteristics of pancreatic cancer but also provide a practical approach to tackle this disease.


Assuntos
Anticorpos Monoclonais/farmacologia , Quimiocinas/metabolismo , Interleucina-17/imunologia , Interleucina-17/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Receptores de Interleucina-17/metabolismo , Animais , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-17/genética , Masculino , Camundongos SCID , Pessoa de Meia-Idade , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/mortalidade , Receptores de Interleucina-17/genética , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Am J Respir Crit Care Med ; 190(6): 675-87, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25152164

RESUMO

RATIONALE: Metabolic alterations contribute to cancer development and progression. However, the molecular mechanisms relating metabolism to cancer metastasis remain largely unknown. OBJECTIVES: To identify a key metabolic enzyme that is aberrantly overexpressed in invasive lung cancer cells and to investigate its functional role and prognostic value in lung cancer. METHODS: The differential expression of metabolic enzymes in noninvasive CL1-0 cells and invasive CL1-5 cells was analyzed by a gene expression microarray. The expression of target genes in clinical specimens from patients with lung cancer was examined by immunohistochemistry. Pharmacologic and gene knockdown/overexpression approaches were used to investigate the function of the target gene during invasion and metastasis in vitro and in vivo. The association between the target gene expression and clinicopathologic parameters was further analyzed. Bioinformatic analyses were used to discover the signaling pathways involved in target gene-regulated invasion and migration. MEASUREMENTS AND MAIN RESULTS: Squalene synthase (SQS) was up-regulated in CL1-5 cells and in the tumor regions of the lung cancer specimens. Loss of function or knockdown of SQS significantly inhibited invasion/migration and metastasis in cell and animal models and vice versa. High expression of SQS was significantly associated with poor prognosis among patients with lung cancer. Mechanistically, SQS contributed to a lipid-raft-localized enrichment of tumor necrosis factor receptor 1 in a cholesterol-dependent manner, which resulted in the enhancement of nuclear factor-κB activation leading to matrix metallopeptidase 1 up-regulation. CONCLUSIONS: Up-regulation of SQS promotes metastasis of lung cancer by enhancing tumor necrosis factor-α receptor 1 and nuclear factor-κB activation and matrix metallopeptidase 1 expression. Targeting SQS may have considerable potential as a novel therapeutic strategy to treat metastatic lung cancer.


Assuntos
Farnesil-Difosfato Farnesiltransferase/metabolismo , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/secundário , Microdomínios da Membrana/metabolismo , Invasividade Neoplásica/fisiopatologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Animais , Linhagem Celular Tumoral , Colesterol/biossíntese , Modelos Animais de Doenças , Farnesil-Difosfato Farnesiltransferase/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Metaloproteinase 1 da Matriz/metabolismo , Prognóstico , Regulação para Cima
9.
EMBO Mol Med ; 5(8): 1165-79, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23828861

RESUMO

Elevated oxidative stress is closely associated with obesity. Emerging evidence shows that instead of being a consequence of obesity, oxidative stress may also contribute to fat formation. Nonselenocysteine-containing phospholipid hydroperoxide glutathione peroxidase (NPGPx) is a conserved oxidative stress sensor/transducer and deficiency of NPGPx causes accumulation of reactive oxygen species (ROS). In this communication, we show that NPGPx was highly expressed in preadipocytes of adipose tissue. Deficiency of NPGPx promoted preadipocytes to differentiate to adipocytes via ROS-dependent dimerization of protein kinase A regulatory subunits and activation of CCAAT/enhancer-binding protein beta (C/EBPß). This enhanced adipogenesis was alleviated by antioxidant N-acetylcysteine (NAC). Consistently, NPGPx-deficient mice exhibited markedly increased fat mass and adipocyte hypertrophy, while treatment with NAC ablated these phenotypes. Furthermore, single nucleotide polymorphisms (SNPs) in human NPGPx gene, which correlated with lower NPGPx expression level in adipose tissue, were associated with higher body mass index (BMI) in several independent human populations. These results indicate that NPGPx protects against fat accumulation in mice and human via modulating ROS, and highlight the importance of targeting redox homeostasis in obesity management.


Assuntos
Proteínas de Transporte/fisiologia , Regulação da Expressão Gênica , Obesidade/genética , Estresse Oxidativo , Peroxidases/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Adipócitos/citologia , Animais , Antioxidantes/metabolismo , Índice de Massa Corporal , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteínas de Transporte/genética , Feminino , Glutationa Peroxidase , Humanos , Masculino , Camundongos , Camundongos Knockout , Obesidade/metabolismo , Peroxidases/genética , Fenótipo , Polimorfismo de Nucleotídeo Único
10.
Proc Natl Acad Sci U S A ; 110(30): 12331-6, 2013 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-23836662

RESUMO

The circadian clock gene Period2 (PER2) has been suggested to be a tumor suppressor. However, detailed mechanistic evidence has not been provided to support this hypothesis. We found that loss of PER2 enhanced invasion and activated expression of epithelial-mesenchymal transition (EMT) genes including TWIST1, SLUG, and SNAIL. This finding was corroborated by clinical observation that PER2 down-regulation was associated with poor prognosis in breast cancer patients. We further demonstrated that PER2 served as a transcriptional corepressor, which recruited polycomb proteins EZH2 and SUZ12 as well as HDAC2 to octamer transcription factor 1 (OCT1) (POU2F1) binding sites of the TWIST1 and SLUG promoters to repress expression of these EMT genes. Hypoxia, a condition commonly observed in tumors, caused PER2 degradation and disrupted the PER2 repressor complex, leading to activation of EMT gene expression. This result was further supported by clinical data showing a significant negative correlation between hypoxia and PER2. Thus, our findings clearly demonstrate the tumor suppression function of PER2 and elucidate a pathway by which hypoxia promotes EMT via degradation of PER2.


Assuntos
Neoplasias da Mama/metabolismo , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica/genética , Hipóxia/genética , Transportador 1 de Cátions Orgânicos/fisiologia , Proteínas Circadianas Period/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Humanos , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Regulação para Cima/genética
11.
J Assist Reprod Genet ; 30(8): 1063-72, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23832270

RESUMO

PURPOSE: Previous studies reported that patients with endometriosis had excess nitric oxide (NO) in the reproductive tract and poor embryo development in IVF cycles. This study aims to elucidate the effects of NO on early embryo development. METHODS: Zygotes from superovulated B6CBF1 mice were cultured to blastocysts in a variety of media. Sodium nitroprusside (SNP) and N(G)-nitro-L-arginine (LNA) were added to the culture medium as a NO donor and a NO synthase inhibitor, respectively. The localization and fluorescence intensity of S-nitrosylated (SNO) proteins within 2-cell stage embryos were analyzed with confocal microscopy. Apoptosis and ATP production in the blastocysts were measured. RESULT(S): Subsequent to NO exposure, the SNO proteins mainly colocalized with the mitochondria and endoplasmic reticulum and the intensity of SNO proteins increased. The addition of a quanylate cyclase inhibitor and a cyclic GMP mimic agent induced nonsignificant changes in SNO proteins, whereas addition of a superoxide scavenger or a reduced form of glutathione rescued the embryos from the effects of NO. However, superoxide scavenger supplementation resulted in decreased blastocyst ATP production. CONCLUSION(S): Elevated NO exerts deleterious effects on embryo development, possibly through protein S-nitrosylation in the mitochondria and endoplasmic reticulum. Including glutathione as a component in the culture medium might counteract this effect.


Assuntos
Apoptose , Blastocisto/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Óxido Nítrico/toxicidade , Animais , Blastocisto/citologia , Blastocisto/ultraestrutura , Técnicas de Cultura Embrionária , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia
12.
Nucleic Acids Res ; 41(3): 1533-43, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23241391

RESUMO

We reported that non-targeting siRNA (NT-siRNA) stress induces non-selenocysteine containing phospholipid hydroperoxide glutathione peroxidase (NPGPx) expression to cooperate with exoribonuclease XRN2 for releasing the stress [Wei,P.C., Lo,W.T., Su,M.I., Shew,J.Y. and Lee,W.H. (2011) Non-targeting siRNA induces NPGPx expression to cooperate with exoribonuclease XRN2 for releasing the stress. Nucleic Acids Res., 40, 323-332]. However, how NT-siRNA stress inducing NPGPx expression remains elusive. In this communication, we showed that the proximal promoter of NPGPx contained a mixed G-quadruplex (G4) structure, and disrupting the structure diminished NT-siRNA induced NPGPx promoter activity. We also demonstrated that nucleolin (NCL) specifically bonded to the G4-containing sequences to replace the originally bound Sp1 at the NPGPx promoter on NT-siRNA stress. Consistently, overexpression of NCL further increased NPGPx promoter activity, whereas depletion of NCL desensitized NPGPx promoter to NT-siRNA stress. These results suggest that the cis-element with mixed G4 structure at the NPGPx promoter plays an essential role for its transactivation mediated by NCL to release cells from NT-siRNA stress.


Assuntos
Quadruplex G , Peroxidases/genética , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno , Proteínas de Ligação a RNA/metabolismo , Estresse Fisiológico/genética , Ativação Transcricional , Sítios de Ligação , Linhagem Celular , Sequência Rica em GC , Humanos , Peroxidases/metabolismo , Fator de Transcrição Sp1/metabolismo , Regulação para Cima , Nucleolina
13.
Mol Cell ; 48(5): 747-59, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23123197

RESUMO

NPGPx is a member of the glutathione peroxidase (GPx) family; however, it lacks GPx enzymatic activity due to the absence of a critical selenocysteine residue, rendering its function an enigma. Here, we show that NPGPx is a newly identified stress sensor that transmits oxidative stress signals by forming the disulfide bond between its Cys57 and Cys86 residues. This oxidized form of NPGPx binds to glucose-regulated protein (GRP)78 and forms covalent bonding intermediates between Cys86 of NPGPx and Cys41/Cys420 of GRP78. Subsequently, the formation of the disulfide bond between Cys41 and Cys420 of GRP78 enhances its chaperone activity. NPGPx-deficient cells display increased reactive oxygen species, accumulated misfolded proteins, and impaired GRP78 chaperone activity. Complete loss of NPGPx in animals causes systemic oxidative stress, increases carcinogenesis, and shortens life span. These results suggest that NPGPx is essential for releasing excessive ER stress by enhancing GRP78 chaperone activity to maintain physiological homeostasis.


Assuntos
Proteínas de Transporte/metabolismo , Estresse do Retículo Endoplasmático , Proteínas de Choque Térmico/metabolismo , Estresse Oxidativo , Peroxidases/metabolismo , Deficiências na Proteostase/enzimologia , Transdução de Sinais , Animais , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Cisteína , Dano ao DNA , Dissulfetos/metabolismo , Relação Dose-Resposta a Droga , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/genética , Fibroblastos/enzimologia , Fibroblastos/patologia , Glutationa Peroxidase , Proteínas de Choque Térmico/genética , Homeostase , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Sítio-Dirigida , Mutação , Oxidantes/farmacologia , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Peroxidases/genética , Ligação Proteica , Dobramento de Proteína , Deficiências na Proteostase/genética , Deficiências na Proteostase/patologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Tempo , Transfecção
14.
Cancer Res ; 72(18): 4652-61, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22826604

RESUMO

Tumor microenvironment plays a critical role in regulating tumor progression by secreting factors that mediate cancer cell growth. Stromal fibroblasts can promote tumor growth through paracrine factors; however, restraint of malignant carcinoma progression by the microenvironment also has been observed. The mechanisms that underlie this paradox remain unknown. Here, we report that the tumorigenic potential of breast cancer cells is determined by an interaction between the Robo1 receptor and its ligand Slit2, which is secreted by stromal fibroblasts. The presence of an active Slit2/Robo1 signal blocks the translocation of ß-catenin into nucleus, leading to downregulation of c-myc and cyclin D1 via the phosphoinositide 3-kinase (PI3K)/Akt pathway. Clinically, high Robo1 expression in the breast cancer cells correlates with increased survival in patients with breast cancer, and low Slit2 expression in the stromal fibroblasts is associated with lymph node metastasis. Together, our findings explain how a specific tumor microenvironment can restrain a given type of cancer cell from progression and show that both stromal fibroblasts and tumor cell heterogeneity affect breast cancer outcomes.


Assuntos
Neoplasias da Mama/metabolismo , Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais/fisiologia , Microambiente Tumoral/fisiologia , Animais , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , beta Catenina/metabolismo , Proteínas Roundabout
15.
PLoS One ; 7(4): e35128, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22514714

RESUMO

Microenvironment plays an important role in cancer development. We have reported that the cancer-associated stromal cells exhibit phenotypic and functional changes compared to stromal cells neighboring to normal tissues. However, the molecular mechanisms as well as the maintenance of these changes remain elusive. Here we showed that through co-culture with breast cancer cells for at least three to four passages, breast normal tissue-associated fibroblasts (NAFs) gained persistent activity for promoting cancer cell invasion, partly via up-regulating ADAM metallopeptidase with thrombospondin type 1 motif, 1 (ADAMTS1). Furthermore, we demonstrated that the DNA methylation pattern in the ADAMTS1 promoter has no alteration. Instead, the loss of EZH2 binding to the ADAMTS1 promoter and the resulting decrease of promoter-associated histone H3K27 methylation may account for the up-regulation of ADAMTS1. Importantly, the lack of EZH2 binding and the H3K27 methylation on the ADAMTS1 promoter were sustained in cancer cell-precocultured NAFs after removal of cancer cells. These results suggest that cancer cells are capable of inducing stromal fibroblasts to secrete ADAMTS1 persistently for their invasion and the effect is epigenetically inheritable.


Assuntos
Proteínas ADAM/metabolismo , Epigênese Genética/genética , Fibroblastos/metabolismo , Proteínas ADAM/genética , Proteína ADAMTS1 , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Imunoprecipitação da Cromatina , Técnicas de Cocultura , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunoprecipitação , Metástase Neoplásica/genética , Proteoglicanas/genética , Proteoglicanas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
16.
Nucleic Acids Res ; 40(1): 323-32, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21908404

RESUMO

Short interfering RNAs (siRNAs) target specific mRNAs for their degradation mediated by RNA-induced silencing complex (RISC). Persistent activation of siRNA-RISC frequently leads to non-targeting toxicity. However, how cells mediate this stress remains elusive. In this communication, we found that the presence of non-targeting siRNA selectively induced the expression of an endoplasmic reticulum (ER)-resident protein, non-selenocysteine containing phospholipid hydroperoxide glutathione peroxidase (NPGPx), but not other ER-stress proteins including GRP78, Calnexin and XBP1. Cells suffering from constant non-targeting siRNA stress grew slower and prolonged G1 phase, while NPGPx-depleted cells accumulated mature non-targeting siRNA and underwent apoptosis. Upon the stress, NPGPx covalently bound to exoribonuclease XRN2, facilitating XRN2 to remove accumulated non-targeting siRNA. These results suggest that NPGPx serves as a novel responder to non-targeting siRNA-induced stress in facilitating XRN2 to release the non-targeting siRNA accumulation.


Assuntos
Exorribonucleases/metabolismo , Glutationa Peroxidase/metabolismo , Peroxidases/metabolismo , RNA Interferente Pequeno/metabolismo , Estresse Fisiológico , Animais , Apoptose , Dano ao DNA , Chaperona BiP do Retículo Endoplasmático , Exorribonucleases/fisiologia , Fase G1 , Glutationa Peroxidase/biossíntese , Glutationa Peroxidase/genética , Humanos , Camundongos , Peroxidases/biossíntese , Peroxidases/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Estresse Fisiológico/genética
17.
PLoS One ; 6(1): e15313, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-21249190

RESUMO

It has been well documented that microenvironment consisting of stroma affects breast cancer progression. However, the mechanisms by which cancer cells and fibroblasts, the major cell type in stroma, interact with each other during tumor development remains to be elucidated. Here, we show that the human cancer-associated fibroblasts (CAFs) had higher activity in enhancing breast tumorigenecity compared to the normal tissue-associated fibroblasts (NAFs) isolated from the same patients. The expression level of hepatocyte growth factor (HGF) in these fibroblasts was positively correlated with their ability to enhance breast tumorigenesis in mice. Deprivation of HGF using a neutralizing antibody reduced CAF-mediated colony formation of human breast cancer cells, indicating that CAFs enhanced cancer cell colony formation mainly through HGF secretion. Co-culture with human breast cancer MDA-MB-468 cells in a transwell system enhanced NAFs to secret HGF as well as promote tumorigenecity. The newly gained ability of these "educated" NAFs became irreversible after continuing this process till fourth passage. These results suggested that breast cancer cells could alter the nature of its surrounding fibroblasts to secrete HGF to support its own progression through paracrine signaling.


Assuntos
Neoplasias da Mama/patologia , Transformação Celular Neoplásica/metabolismo , Fibroblastos/patologia , Fator de Crescimento de Hepatócito/fisiologia , Comunicação Parácrina , Microambiente Tumoral/fisiologia , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura , Progressão da Doença , Feminino , Fibroblastos/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Neoplasias Mamárias Animais/patologia , Camundongos
18.
Head Neck ; 30(12): 1575-85, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18798303

RESUMO

BACKGROUND: Inhibitors of apoptosis proteins (IAPs), which counteract apoptosis by potently inhibiting caspase activation, are promising targets of new anti-tumor therapy. However, their roles in the pathogenesis of nasopharyngeal carcinoma (NPC), an Epstein-Barr virus (EBV)-associated carcinoma, are not fully understood. Herein, we investigated the expression and regulation of IAPs in NPC. METHODS AND RESULTS: Using real-time quantitative polymerase chain reaction (PCR) analysis, we found that among the IAPs family only the transcription of survivin, HIAP-1, and HIAP-2 was consistently up-regulated in NPC and metastatic NPC tissues. Immunohistochemical staining showed that their proteins were more predominantly expressed in tumor cells nests. Noteworthy, these IAPs were upregulated by interleukin-1 alpha stimulation or EBV infection, and subsequently resulted in triggering rapid proliferation of NPC verified by strong Ki-67 staining. CONCLUSION: Survivin, HIAP-1, and HIAP-2 were distinctly upregulated in NPC, suggesting they may play significant roles in NPC tumorigenesis and serve as tumor markers with prognostic and therapeutic implications.


Assuntos
Carcinoma de Células Escamosas/genética , Infecções por Vírus Epstein-Barr/genética , Herpesvirus Humano 4/genética , Proteínas Inibidoras de Apoptose/genética , Proteínas Associadas aos Microtúbulos/genética , Neoplasias Nasofaríngeas/genética , Apoptose , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/virologia , Caspases/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Humanos , Interleucina-1alfa/genética , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/virologia , Reação em Cadeia da Polimerase , Survivina
19.
Cancer Epidemiol Biomarkers Prev ; 17(3): 535-42, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18349270

RESUMO

Previous studies have identified that the expression of UK114 is tissue specific and the protein has been found to be most abundant in liver and kidney. However, the expression of UK114 in human hepatocellular carcinoma and its relationship to differentiation and transformation of hepatocellular carcinoma have not been studied. In this study, the expression of UK114 in human hepatocellular carcinoma was examined by Northern and Western blot analyses. We found that UK114 was significantly down-regulated in most of hepatocellular carcinoma tissues compared with adjacent nontumor tissues (72.7%) at both mRNA and protein levels. We looked into the possibility that this decreased expression of UK114 in the hepatocellular carcinoma tissues may play a role in the differentiation or tumorigenicity of hepatocellular carcinoma. Immunohistochemical staining showed that the reduced expression of UK114 in hepatocellular carcinoma tissues was correlated with the tumor differentiation status as graded by the Edmondson-Steiner classification. On the other hand, overexpression of UK114 was not able to suppress the proliferation of human hepatoma cells and tumorigenicity in nude mice. These results suggest that UK114 does not seem to act as a tumor suppressor gene; however, it may useful as a biomarker that will assist in the grading of the differentiation status of hepatocellular carcinoma samples.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proteínas de Choque Térmico/metabolismo , Neoplasias Hepáticas/metabolismo , Ribonucleases/metabolismo , Biomarcadores Tumorais/metabolismo , Northern Blotting , Western Blotting , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Distribuição de Qui-Quadrado , Regulação para Baixo , Humanos , Técnicas Imunoenzimáticas , Rim/metabolismo , Fígado/metabolismo , Neoplasias Hepáticas/patologia , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA