Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Front Public Health ; 12: 1333910, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38439751

RESUMO

Background: Metabolic syndrome (MetS) is on the rise in developing countries and is characterized by a series of indications of metabolic disturbance. However, the prevalence of MetS varies under different definitions. The study aimed to compare five definitions of MetS in the China adult population, to explore their prevalence, characteristics and agreement. Methods: The data for the retrospective study came from the China Health and Retirement Longitudinal Study (CHARLS), consisting of 9,588 participants (≥45). MetS definitions from International Diabetes Federation (IDF) (2006), National Cholesterol Education Program Adult Treatment Panel III (ATPIII) (2005), National Cholesterol Education Program Adult Treatment Panel III (ATPIII) (2001), Chinese Diabetes society (CDS) (2004) and the World Health Organization (WHO) (1999). We used binary and multivariable logistic analysis to explore factors connected with MetS. Results: The five definitions of MetS led to different prevalence of MetS:34.52% by IDF (2006), 38.63% by ATP (2005), 25.94% by ATP (2001), 26.31% by CDS (2004), 21.57% by WHO (1999). According to the definition of IDF (2006) (22.32% vs. 45.06%), ATPIII (2005) definition (27.99% vs. 47.82%), ATPIII (2001) definition (15.37% vs. 35.07%), CDS (2004) definition (19.96% vs. 31.80%), and WHO (1999) definition (17.44% vs. 25.14%), the prevalence of MetS in men was low but in women was high. The agreement between the five definitions for men was good except for the IDF (2006) definition and ATPIII (2001) definition (kappa = 0.51), with kappa values from 0.64 to 0.85. For women, the agreement between the five definitions was good ranging from 0.67 to 0.95, however, except for the definition of CDS (2004) and the definition of IDF (2006) (kappa = 0.44), the definition of WHO (1999) and the definition of IDF (2006) (kappa = 0.55), and the definition of WHO (1999) and the definition of ATPIII (2005) (kappa = 0.54). Binary logistic analysis indicated that although the impact and relevance varied by sex and definition, age, education, marital status, current residence, current smoking, alcohol using, taking activities and number of chronic diseases were factors connected to MetS. Conclusion: the prevalence and characteristics of the five definitions of MetS are different in the Chinese population. Therefore, it is vital to use the same definition for a country to diagnose MetS. On the other side, a lower prevalence in men than in women and the consistency of five MetS definitions are good in men but relatively poor in women.


Assuntos
Diabetes Mellitus , Síndrome Metabólica , Adulto , Masculino , Feminino , Humanos , Síndrome Metabólica/diagnóstico , Síndrome Metabólica/epidemiologia , Estudos Retrospectivos , Estudos Longitudinais , Prevalência , China/epidemiologia , Colesterol , Trifosfato de Adenosina
2.
Biochem Biophys Res Commun ; 629: 121-127, 2022 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-36116374

RESUMO

CD82, a tetraspanin superfamily member, has been identified to be glycosylated at three specific residues (Asn129, Asn157, and Asn198). However, CD82 post-translational modification and its effect on colorectal cancer (CRC) metastasis remain unclear. Here, we constructed various deficient mutants of CD82 N-glycosylation in SW620 cells and demonstrated that the Asn157 site is necessary for CD82 glycosylation in CRC cells migration and LN-dependent adhesion in vitro. Furthermore, we found that CD82 N-glycosylation at the Asn157 site leads to lower expression levels of vimentin and claudin-1 but higher expression levels of E-cadherin, which are the EMT markers; also, there are lower expression levels of phospho-GSK3ß and less ß-catenin transportation to the nucleus. These findings suggest that CD82 N-glycosylation at the Asn157 site inhibits EMT by down-regulating the Wnt/ß-catenin pathway. Moreover, we reported that CD82 with N-glycosylation at a single site of the Asn157 reduces lung metastases in vivo. The results indicate that N-glycosylation of CD82 at the Asn157 site regulates CRC metastasis and adhesion. These observations suggest that the N-glycosylation of CD82 might be a potential therapeutic target for CRC.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Claudina-1/metabolismo , Neoplasias do Colo/genética , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Glicosilação , Humanos , Proteína Kangai-1/genética , Proteína Kangai-1/metabolismo , Vimentina/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
4.
Biochem Biophys Res Commun ; 557: 110-116, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33862453

RESUMO

The peptide mimicking small extracellular loop of CD82/KAI1 has been reported to inhibit tumor cell migration and metastasis. This provides an evidence that small extracellular loop domain should be important for the function of CD82/KAI1. In this paper, to investigate the structure basis for the function of EC1 mimic peptide, we systematically analyzed the effects of each amino acid residue in EC1 mimic peptide on its bioactivity. We found that the interfering with the folding of secondary structure with proline, a potent breaker of secondary structure, completely abolished the migration and metastasis-inhibitory activity of EC1 mimic peptide. This means that the bioactivity of EC1 mimic peptide was conformation-dependent. Next, we substitute with proline for amino acid residues in the small extracellular ring region of CD82/KAI1 by the site-specific mutations to disrupting secondary structure and detected its effect on the function of CD82/KAI1. The results showed that the disturbing the secondary structure of small extracellular ring completely abolished the migration and metastasis-inhibitory activity of CD82/KAI1. These results further provide direct evidence that the small extracellular ring is an important function region of CD82/KAI1.


Assuntos
Neoplasias da Mama/metabolismo , Proteína Kangai-1/metabolismo , Neoplasias Pulmonares/metabolismo , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/prevenção & controle , Linhagem Celular Tumoral , Movimento Celular , Feminino , Genes Supressores de Tumor , Humanos , Proteína Kangai-1/química , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Prolina/química , Prolina/metabolismo , Domínios Proteicos , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Cancer Res Clin Oncol ; 147(7): 1927-1934, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33811273

RESUMO

BACKGROUND: Tetraspanin KAI1/CD82, a tumor metastasis suppressor, has emerged as a promising molecular target for the management of metastatic disease. However, the peptide mimicking small extracellular ring domain (EC1) of CD82 has not been fully investigated for the function of inhibiting cell migration in vitro and tumor metastasis in vivo. METHODS: Different cancer cells were treated with EC1 mimic peptide in order to detect migration and invasion by the healing assay and transwell. Cell aggregation and adhesion assays were used to investigate the function of homotypic cell-cell aggregation and adhesion to tissue culture plates. Then, we established syngeneic and xenograft animal models to assess the metastasis inhibitory effect of EC1 mimic peptide in vivo. RESULTS: In vitro studies, the EC1 mimic peptide had been showed to promote homotypic cell-cell aggregation, suppress cell migration, invasion and adherence in multiple tumor cell types. In vivo metastasis assays, the EC1 mimic peptide could strongly inhibit the pulmonary metastasis of LCC in syngeneic mice model and SW620 and H1299 in xenograft mice model. CONCLUSION: This novel finding will improve our understanding of the mechanism by which CD82 inhibits metastasis, and suggests that EC1 mimic peptide may be a promising candidate for developing anti-metastasis drugs.


Assuntos
Movimento Celular , Proteína Kangai-1/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Fragmentos de Peptídeos/farmacologia , Animais , Apoptose , Proliferação de Células , Humanos , Técnicas In Vitro , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Nus , Metástase Neoplásica , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Mol Med Rep ; 22(5): 3994-4002, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33000220

RESUMO

Previous studies have shown that (GM3), a ganglioside, suppresses hepatoma cell motility and migration by inhibiting phosphorylation of EGFR and the activity of the PI3K/AKT signaling pathway. Therefore, the aim of the present study was to investigate whether the combined treatment of CD82 with gangliosides can exert a synergistic inhibitory effect on cell motility and migration. Epidermal growth factor receptor (EGFR) signaling was studied for its role in the mechanism through which CD82 and gangliosides synergistically inhibit the motility and migration of SW620 human colon adenocarcinoma cells. GM3 and/or GM2 treatment, and/or overexpression of CD82 was performed in SW620 cells. High-performance thin layer chromatography, reverse transcription-quantitative PCR, western blotting and flow cytometry assays were used to confirm the content changes of GM2, GM3 and CD82. In addition, the phosphorylation of EGFR, MAPK and Akt were evaluated by western blot analysis. SW620 cell motility was investigated using wound healing analysis and chemotaxis migration assay. The combination of GM3 and GM2 with CD82 was found to markedly suppress EGF-stimulated SW620 cell motility compared with the individual factors or combination of GM2 or GM3 with CD82 by inhibiting the phosphorylation of EGFR. The results suggested that CD82 in combination with either GM2 or GM3 can exert a synergistic inhibitory effect on cell motility and migration; however, the synergistic mechanisms elicited by GM2 or GM3 with CD82 differ.


Assuntos
Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Neoplasias do Colo/metabolismo , Gangliosídeo G(M2)/farmacologia , Gangliosídeo G(M3)/farmacologia , Proteína Kangai-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tirosina/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Receptores ErbB/metabolismo , Humanos , Proteína Kangai-1/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Transfecção
7.
Biochem Biophys Res Commun ; 533(3): 338-345, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-32958256

RESUMO

We have previously demonstrated that the peptide mimicking small extracellular ring domain of CD82 (CD82EC1-mP) could inhibit tumor cell motility and metastasis. However, its acting mechanism is not understood. Here, we reported that the cell motility-inhibitory function of CD82EC1-mP was involved in the downregulation of epithelial-mesenchymal transition (EMT). Both vimentin and E-cadherin are EMT makers. We found that CD82EC1-mP could inhibit the expression of vimentin, but promot the expression of E-cadherin, suggesting that CD82EC1-mP suppressed EMT. Hippo/YAP and Wnt/ß-catenin are both key signal pathways that regulate the EMT process. The futher studies showed that CD82EC1-mP couled activate GSK3ß, promote the phosphorylation of ß-catenin, and inhibit the ß-catenin nuclear location. Moreover, CD82EC1-mP couled activate Hipoo kinase cascade, promote the phosphorylation of YAP, and inhibit the YAP nuclear location. These results suggested that CD82EC1-mP inhibited invation and matestasis via inhibiting EMT through downregulating Wnt pathway and upregulating Hippo pathway.


Assuntos
Antineoplásicos/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Proteína Kangai-1/genética , Peptídeos/farmacologia , Proteínas Serina-Treonina Quinases/genética , Via de Sinalização Wnt/efeitos dos fármacos , Células A549 , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Antineoplásicos/síntese química , Caderinas/agonistas , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Células Hep G2 , Via de Sinalização Hippo , Humanos , Proteína Kangai-1/antagonistas & inibidores , Proteína Kangai-1/química , Proteína Kangai-1/metabolismo , Mimetismo Molecular , Células PC-3 , Peptídeos/síntese química , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vimentina/antagonistas & inibidores , Vimentina/genética , Vimentina/metabolismo , Proteínas de Sinalização YAP , beta Catenina/genética , beta Catenina/metabolismo
8.
Int J Mol Med ; 45(3): 939-946, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31985021

RESUMO

Acute kidney injury (AKI) is characterized by abrupt kidney dysfunction. It results in remote organ dysfunction, including the brain. The underlying mechanism of the kidney­brain axis in AKI and effective protective approaches remain unknown. The present study aimed to investigate the potential protective effect of ginsenoside (GS) on AKI induced by glycerol in rats. Kidney function was initially assessed by blood urea nitrogen (BUN) and creatinine (Cre) tests, and was identified to be severely impaired following glycerol treatment, based on significant increases in BUN and Cre levels observed. Severe extensive necrosis of the majority of the renal tubules was observed by hematoxylin and eosin staining, additionally confirming that glycerol induced AKI. GS was identified to ameliorate the impairment of kidney function in the context of AKI. Further investigation of the mechanism revealed that GS may induce protection against oxidative stress via a kidney­brain axis. Furthermore, GS improved the activation of hypoxia­inducible factor 1α (HIF­1α) and vascular endothelial growth factor A (VEGF­A) in the hypothalamus response to AKI, and in the kidney tissues. The protective effect of GS in AKI may be associated with the interaction between the kidney and the brain. Taken together, these results suggested that GS was involved in the protective effects against AKI by decreasing oxidative damage to the kidney and brain, and by upregulating HIF­1α and VEGF­A levels in the kidney­brain axis.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Ginsenosídeos/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Nitrogênio da Ureia Sanguínea , Western Blotting , Creatinina/metabolismo , Imuno-Histoquímica , Rim/efeitos dos fármacos , Rim/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
9.
Mol Carcinog ; 58(5): 674-685, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30604894

RESUMO

CD82, a member of the tetraspanin superfamily, has been proposed to exert its activity via tetra-transmembrane protein enriched microdomains (TEMs) in exosomes. The present study aimed to explore the potential of the exosome protein CD82 in diagnosing breast cancers of all stages and various histological subtypes in patients. The results strongly suggest that CD82 expression in breast cancer tissue was significantly lower than that in healthy and benign breast disease tissues. There was a significant negative correlation between CD82 expression in tissues and CD82 content in exosomes, which indicated that CD82 expression was redistributed from tissues to the blood with the development and metastasis of breast cancer.


Assuntos
Adenocarcinoma Mucinoso/diagnóstico , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/diagnóstico , Carcinoma Ductal de Mama/diagnóstico , Carcinoma Lobular/diagnóstico , Exossomos/metabolismo , Proteína Kangai-1/metabolismo , Adenocarcinoma Mucinoso/metabolismo , Adulto , Idoso , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Lobular/metabolismo , Estudos de Casos e Controles , Estudos de Viabilidade , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Medicina de Precisão , Prognóstico
10.
Biochem Biophys Res Commun ; 503(4): 2206-2211, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-29953857

RESUMO

Within the extracellular domains of metastasis suppressor CD82, the large extracellular loop (EC2) has received much of the attention and its structure and function have been studied in detail. However, little attention has been given to the small extracellular loop (EC1 domain). To investigate the function role of EC1 in metastasis suppression of CD82, the peptide mimicking EC1 amino acid sequence (EC1-mP) was synthesized and its effect on cancer cells behavior was examined. Here, we reported that EC1-mP strongly inhibited cancer cell migration in vitro, attnuated the ability of cancer cells adhesion to fibronectin, and induced the apoptosis. Furthermore, the EC1-mP was showed to supprese the expressions of integrins α5 and ß1, as well as decreased the phosphorylation of FAK and expression of ILK in SW620 cells. Taken together, these results demonstrate that this small peptide has the functional role of CD82 intact molecule. This novel finding will improve our understanding of the mechanism by which CD82 inhibits metastasis, and suggested that EC1 mimic peptide may be a promising candidate for developing anti-metastasis drugs.


Assuntos
Proteína Kangai-1/genética , Peptídeos/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Humanos , Integrina alfa5/efeitos dos fármacos , Integrina beta1/efeitos dos fármacos , Mimetismo Molecular , Metástase Neoplásica , Domínios Proteicos , Transdução de Sinais/efeitos dos fármacos
11.
Mol Med Rep ; 11(4): 2959-66, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25503644

RESUMO

The ganglioside GM3 exerts its different effects via various growth factor receptors. The present study investigated and comparatively analyzed the opposing effects exerted by GM3 on the migration of mouse hepatocellular carcinoma Hepa1­6 cells via epidermal growth factor receptor (EGFR) and hepatocyte growth factor receptor (HGFR/cMet). The results demonstrated that GM3 inhibited EGF­stimulated motility, but promoted HGF­stimulated motility of the Hepa1­6 cells via phosphatidylinositol 3­kinase/Akt­mediated migration signaling. It is well established that the main cytokines modulating cell proliferation, invasion and metastasis are different in different types of tumor. This difference may, at least in part, explain why GM3 exerted its actions in a tumor­type specific manner.


Assuntos
Movimento Celular/efeitos dos fármacos , Receptores ErbB/metabolismo , Gangliosídeo G(M3)/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/farmacologia , Gangliosídeo G(M3)/biossíntese , Fator de Crescimento de Hepatócito/farmacologia , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Sialiltransferases/genética
12.
Mol Neurobiol ; 49(2): 645-57, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24068615

RESUMO

In Lafora disease (LD), the deficiency of either EPM2A or NHLRC1, the genes encoding the phosphatase laforin and E3 ligase, respectively, causes massive accumulation of less-branched glycogen inclusions, known as Lafora bodies, also called polyglucosan bodies (PBs), in several types of cells including neurons. The biochemical mechanism underlying the PB accumulation, however, remains undefined. We recently demonstrated that laforin is a phosphatase of muscle glycogen synthase (GS1) in PBs, and that laforin recruits malin, together reducing PBs. We show here that accomplishment of PB degradation requires a protein assembly consisting of at least four key enzymes: laforin and malin in a complex, and the glycogenolytic enzymes, glycogen debranching enzyme 1 (AGL1) and brain isoform glycogen phosphorylase (GPBB). Once GS1-synthesized polyglucosan accumulates into PBs, laforin recruits malin to the PBs where laforin dephosphorylates, and malin degrades the GS1 in concert with GPBB and AGL1, resulting in a breakdown of polyglucosan. Without fountional laforin-malin complex assembled on PBs, GPBB and AGL1 together are unable to efficiently breakdown polyglucosan. All these events take place on PBs and in cytoplasm. Deficiency of each of the four enzymes causes PB accumulation in the cytoplasm of affected cells. Demonstration of the molecular mechanisms underlying PB degradation lays a substantial biochemical foundation that may lead to understanding how PB metabolizes and why mutations of either EPM2A or NHLRC1 in humans cause LD. Mutations in AGL1 or GPBB may cause diseases related to PB accumulation.


Assuntos
Encéfalo/enzimologia , Proteínas de Transporte/metabolismo , Glucanos/metabolismo , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Glicogênio Fosforilase/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Animais , Proteínas de Transporte/análise , Linhagem Celular Tumoral , Glucanos/análise , Sistema da Enzima Desramificadora do Glicogênio/análise , Glicogênio Fosforilase/análise , Células HEK293 , Humanos , Isoenzimas/análise , Isoenzimas/metabolismo , Doença de Lafora/metabolismo , Doença de Lafora/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Tirosina Fosfatases não Receptoras/análise , Ubiquitina-Proteína Ligases
13.
PLoS One ; 8(10): e77387, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24143228

RESUMO

Plasmalogens play multiple roles in the structures of biological membranes, cell membrane lipid homeostasis and human diseases. We report the isolation and identification of choline plasmalogens (ChoPlas) from swine liver by high performance thin layer chromatography (HPTLC) and high performance liquid chromatography (HPLC)/MS. The growth and viability of hepatoma cells (CBRH7919, HepG2 and SMMC7721) was determined following ChoPlas treatment comparing with that of human normal immortal cell lines (HL7702). Result indicated that ChoPlas inhibited hepatoma cell proliferation with an optimal concentration and time of 25 µmol/L and 24 h. To better understand the mechanism of the ChoPlas-induced inhibition of hepatoma cell proliferation, Caveolin-1 and PI3K/Akt pathway signals, including total Akt, phospho-Akt(pAkt) and Bcl-2 expression in CBRH7919 cells, were determined by western blot. ChoPlas treatment increased Caveolin-1 expression and reduced the expression of phospho-Akt (pAkt) and Bcl-2, downstream targets of the PI3K/Akt pathway. Further cell cycle analysis showed that ChoPlas treatment induced G1 and G1/S phase transition cell cycle arrest. The expression of essential cell cycle regulatory proteins involved in the G1 and G1/S phase transitions, cyclin D, CDK4, cyclin E and CDK2, were also analyzed by western blot. ChoPlas reduced CDK4, cyclin E and CDK2 expression. Taken together, the results indicate that swine liver-derived natural ChoPlas inhibits hepatoma cell proliferation associated with Caveolin-1 and PI3K/Akt signals.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/patologia , Fígado/química , Plasmalogênios/isolamento & purificação , Plasmalogênios/farmacologia , Transdução de Sinais/efeitos dos fármacos , Suínos , Animais , Carcinoma Hepatocelular/patologia , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Plasmalogênios/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos
14.
Int J Biochem Cell Biol ; 45(11): 2349-58, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23968914

RESUMO

The metastasis suppressor CD82/KAI-1, which is a member of the tetraspanin superfamily, has been proposed to exert its activity together with glycosphingolipids. However, the mechanism of CD82 inhibition has not been fully elucidated. The present study aimed to investigate the synergistic inhibition of cell migration by the tetraspanin CD82 and gangliosides and to correlate this inhibition with activation of epidermal growth factor receptor (EGFR) and hepatocyte growth factor receptor (HGFR/cMet) in Hepa1-6 cell lines, whose motility and migration is stimulated by epidermal growth factor (EGF) and hepatocyte growth factor (HGF) in vitro. We found that Hepa1-6 cells transfected with the CD82 gene exhibited decreased migration in response to EGF and HGF. EGF-stimulated phosphorylation of EGFR at Tyr1173 was inhibited in these cells, which contributed to the attenuation of EGFR. Ectopic expression of CD82 in Hepa1-6 cells inhibited HGF-stimulated tyrosine phosphorylation of cMet at Tyr1313 and Tyr1365 without affecting the expression of cMet. These inhibitory effects were enhanced when CD82 was introduced with Ganglioside GM3 alone or GM2/GM3. Reduction of CD82 expression by RNA interference together with depletion of glycosphingolipids with P4 significantly enhanced cell motility and increased the expression of EGFR and its phosphorylation at Tyr1173 in response to EGF. Increased cell motility and HGF-dependent activation of cMet at Tyr1313 and Tyr1365 resulted from decreased CD82 levels and increased GM3. Furthermore, CD82 expression selectively attenuated EGFR and cMet signalling via phosphatidylinositol 3-kinase/Akt but had no affect on the activity of the MAPK signalling pathway. These results suggest that the synergistic effects of CD82 and GM3 or GM2/GM3 on EGFR expression and phosphorylation and cMet activation are responsible for CD82 inhibition of EGF- and HGF-dependent cell motility and migration of Hepa1-6 cells.


Assuntos
Movimento Celular/efeitos dos fármacos , Receptores ErbB/metabolismo , Gangliosídeos/farmacologia , Proteína Kangai-1/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/farmacologia , Gangliosídeo G(M2)/farmacologia , Gangliosídeo G(M3)/farmacologia , Fator de Crescimento de Hepatócito/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Regulação para Cima/efeitos dos fármacos
15.
Mol Med Rep ; 8(2): 655-61, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23778885

RESUMO

The aim of the present study was to determine the molecular mechanism by which the hepatocyte growth factor (HGF) receptor (cMet) regulates lymphatic metastasis in hepatocellular carcinoma. Mouse hepatoma ascites cell lines with different lymph node metastatic potentials, Hca­F (high metastatic potential) and Hca­P (low metastatic potential), were cultured in vitro. Cells were treated with HGF, fibronectin (FN) and laminin (LN), and the phosphorylated tyrosine residues of cMet and the activities of intracellular phospholipase Cγ/diacylglycerol/protein kinase C (PLCγ/DAG/PKC) and phosphoinositol­3­kinase/protein kinase B (PI3K/AKT) signaling pathways were analyzed comparatively in the two cell lines using western blot analysis and migration assays. Following HGF treatment, the phosphorylation of cMet at Tyr 1313 and 1365 in Hca­F cells was higher, while the phosphorylation of cMet at Tyr 1349 was lower than that in Hca­P. The activity of PLCγ/DAG/PKC was increased in Hca­F cells compared with Hca­P cells, whereas the activity of PI3K/AKT was reduced. After FN treatment, the phosphorylation of cMet at Tyr 1313 and the activity of the PLCγ/DAG/PKC signaling pathway was increased in Hca­F cells compared with Hca­P cells. Following LN treatment, the phosphorylation of cMet at Tyr 1365 and the activity of PLCγ/DAG/PKC was higher in Hca­F cells than in Hca­P cells. Results of the current study indicate that a number of ligands stimulate the phosphorylation of cMet at various tyrosine residues, activating different signaling transduction pathways. In addition, the same ligand was observed to phosphorylate different tyrosine residues on cMet in the two cell lines, as well as activate different intracellular signaling transduction pathways. After cMet is activated, various tyrosine residues are phosphorylated, leading to the activation of the PI3K/AKT and PLCγ/DAG/PKC signaling pathways to different extents in the two cells lines. These results may be important in determining the lymph node metastatic potentials of the two cell lines.


Assuntos
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Fibronectinas/farmacologia , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Linfonodos/patologia , Metástase Linfática , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
Mol Cell Biochem ; 382(1-2): 83-92, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23749170

RESUMO

Ganglioside GM3 plays a well-documented and important role in the regulation of tumor cell proliferation, invasion, and metastasis by modulating tyrosine kinase growth factor receptors. However, the effect of GM3 on the hepatocyte growth factor receptor (HGFR, cMet) has not been fully delineated. In the current study, we investigated how GM3 affects cMet signaling and HGF-stimulated cell motility and migration using three hepatic cancer cell lines of mouse (Hca/A2, Hca/16A3, and Hepa1-6). Decreasing GM3 expression with the use of P4, a specific inhibitor for ganglioside synthesis inhibited the HGF-stimulated phosphorylation of cMet and activity of PI3K/Akt signaling pathway. In contrast, the increased expression of GM3 as a result of adding exogenous GM3 enhanced the HGF-stimulated phosphorylation of cMet and activity of PI3K/Akt signaling pathway. Furthermore, HGF-stimulated cell motility and migration in vitro were inhibited by reduced expression of GM3 and enhanced by increased expression of GM3. All the observations indicate that ganglioside GM3 promotes HGF-stimulated motility of murine hepatoma cell through enhanced phosphorylation of cMet at specific tyrosine sites and PI3K/Akt-mediated migration signaling.


Assuntos
Carcinoma Hepatocelular/patologia , Movimento Celular/efeitos dos fármacos , Gangliosídeo G(M3)/farmacologia , Fator de Crescimento de Hepatócito/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfotirosina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Carcinoma Hepatocelular/enzimologia , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Gangliosídeo G(M2)/farmacologia , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/patologia , Camundongos , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
17.
Mol Neurobiol ; 48(1): 49-61, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23546741

RESUMO

Glycogen, the largest cytosolic macromolecule, is soluble because of intricate construction generating perfect hydrophilic-surfaced spheres. Little is known about neuronal glycogen function and metabolism, though progress is accruing through the neurodegenerative epilepsy Lafora disease (LD) proteins laforin and malin. Neurons in LD exhibit Lafora bodies (LBs), large accumulations of malconstructed insoluble glycogen (polyglucosans). We demonstrated that the laforin-malin complex reduces LBs and protects neuronal cells against endoplasmic reticulum stress-induced apoptosis. We now show that stress induces polyglucosan formation in normal neurons in culture and in the brain. This is mediated by increased glucose-6-phosphate allosterically hyperactivating muscle glycogen synthase (GS1) and is followed by activation of the glycogen digesting enzyme glycogen phosphorylase. In the absence of laforin, stress-induced polyglucosans are undigested and accumulate into massive LBs, and in laforin-deficient mice, stress drastically accelerates LB accumulation and LD. The mechanism through which laforin-malin mediates polyglucosan degradation remains unclear but involves GS1 dephosphorylation by laforin. Our work uncovers the presence of rapid polyglucosan metabolism as part of the normal physiology of neuroprotection. We propose that deficiency in the degradative phase of this metabolism, leading to LB accumulation and resultant seizure predisposition and neurodegeneration, underlies LD.


Assuntos
Progressão da Doença , Fosfatases de Especificidade Dupla/metabolismo , Estresse do Retículo Endoplasmático , Glucanos/metabolismo , Corpos de Inclusão/metabolismo , Doença de Lafora/patologia , Neurônios/patologia , Regulação Alostérica , Animais , Apoptose , Ativação Enzimática , Técnicas de Silenciamento de Genes , Glicogênio/biossíntese , Glicogênio Fosforilase/metabolismo , Glicogênio Sintase/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Hidrólise , Doença de Lafora/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Degeneração Neural/enzimologia , Degeneração Neural/patologia , Neurônios/enzimologia , Fosforilação , Proteínas Tirosina Fosfatases não Receptoras , alfa-Amilases/metabolismo
18.
J Cell Sci ; 124(Pt 21): 3568-80, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22045735

RESUMO

Mutations in sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2) underlie Darier disease (DD), a dominantly inherited skin disorder characterized by loss of keratinocyte adhesion (acantholysis) and abnormal keratinization (dyskeratosis) resulting in characteristic mucocutaneous abnormalities. However, the molecular pathogenic mechanism by which these changes influence keratinocyte adhesion and viability remains unknown. We show here that SERCA2 protein is extremely sensitive to endoplasmic reticulum (ER) stress, which typically results in aggregation and insolubility of the protein. Depletion of ER calcium stores is not necessary for the aggregation but accelerates the progression. Systematic analysis of diverse mutants identical to those found in DD patients demonstrated that the ER stress initiator is the SERCA2 mutant protein itself. These SERCA2 proteins were found to be less soluble, to aggregate and to be more polyubiquitinylated. After transduction into primary human epidermal keratinocytes, mutant SERCA2 aggregates elicited ER stress, caused increased numbers of cells to round up and detach from the culture plate, and induced apoptosis. These mutant induced events were exaggerated by increased ER stress. Furthermore, knockdown SERCA2 in keratinocytes rendered the cells resistant to apoptosis induction. These features of SERCA2 and its mutants establish a mechanistic base to further elucidate the molecular pathogenesis underlying acantholysis and dyskeratosis in DD.


Assuntos
Apoptose , Doença de Darier/enzimologia , Estresse do Retículo Endoplasmático , Queratinócitos/citologia , Mutação , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/química , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Animais , Células Cultivadas , Doença de Darier/genética , Doença de Darier/metabolismo , Doença de Darier/fisiopatologia , Humanos , Queratinócitos/enzimologia , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Solubilidade
19.
IUBMB Life ; 61(8): 846-52, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19517528

RESUMO

We previously established a line of phosphatidylethanolamine N-methyltransferase 2 (pemt2) -stably transfected CBRH-7919 hepatoma cells, and showed that pemt2 over-expression inhibited cell proliferation and induced apoptosis. This study was aimed to further elucidate the cellular mechanisms leading to this apoptosis in these cells. Fatty acid compositions of phosphatidylcholine (PC) in pemt2 over-expressed cells and control cells, and the location of PC synthesized by PEMT2 pathway were analyzed with lipid extraction, high-performance thin layer chromatography, high-performance gas chromatography (HPGC), and [(3)H]-ethanolamine tracing. The effects of pemt2 over-expression on the mitochondrial membrane fluidity, the release of cytochrome C from mitochondria, and the activity of caspases were determined by Western blot. Newly synthesized PC by PEMT2 contained more acyl groups of oleic acid (P < 0.01) and was mainly located in mitochondria; pemt2 over-expression increased the mitochondrial membrane fluidity and the release of cytochrome C from the mitochondria into the cytoplasma, which in turn activated caspase-9 and caspase-3, the key molecules in the mitochondrial apoptotic pathway. We demonstrated that, in rat hepatoma cells, PEMT2-induced apoptosis proceeds through mitochondria.


Assuntos
Apoptose/fisiologia , Membranas Mitocondriais/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Cromatografia Gasosa , Cromatografia em Camada Fina , Citocromos c/metabolismo , Membranas Mitocondriais/efeitos dos fármacos , Fosfatidiletanolamina N-Metiltransferase/farmacologia , Ratos
20.
Mol Cell Biochem ; 331(1-2): 1-7, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19449091

RESUMO

LeY oligosaccharide is stage specifically expressed by the embryo and uterine endometrium, and it plays important roles in embryo implantation. In addition to participating in the recognition and adhesion on fetal-maternal interface, LeY potentially regulates the expression of some implantation-related factors. However, it remains elusive whether it can mediate the involved signaling pathway. In this study, agarose-LeY beads were used to mimic the embryos, and the effects of LeY oligosaccharide on DAG/PKC signaling pathway was studied in human endometrial epithelial cells. Results showed that LeY could significantly trigger the activation of cPKCalpha and cPKCbeta2, and their translocation from the cytosol to the plasma membrane. The cellular DAG content was also upregulated, and the activation of PLCgamma1 was promoted. On the contrary, DAG/PKC signaling pathway was significantly inhibited when anti-LeY antibody was used after confirmation of LeY expression in human endometrial epithelial cells by immunohistochemistry and flow cytometry. These results suggest that LeY oligosaccharide acts as a signal molecule to modulate DAG/PKC signaling pathway.


Assuntos
Diglicerídeos/metabolismo , Endométrio/citologia , Endométrio/enzimologia , Antígenos do Grupo Sanguíneo de Lewis/metabolismo , Proteína Quinase C/metabolismo , Transdução de Sinais , Regulação para Cima , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Feminino , Humanos , Oligossacarídeos/metabolismo , Fosfolipase C gama/metabolismo , Fosforilação , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA