Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 151(3): 370-385, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31063584

RESUMO

Neuronal expression of beta-secretase 1 (BACE1) has been implicated in the progression of Alzheimer's disease. However, the mechanisms that regulate BACE1 expression are unclear. Here, we show that peroxisome proliferator-activated receptor delta (PPARδ) decreases BACE1 expression by up-regulating suppressor of cytokine signaling 1 (SOCS1) in SH-SY5Y neuroblastoma cells. The activation of PPARδ by GW501516, a specific PPARδ agonist, inhibited expression of BACE1. This effect was abrogated by shRNA-mediated knockdown of PPARδ and by treatment with the PPARδ antagonist GSK0660, indicating that PPARδ is involved in GW501516-mediated suppression of BACE1 expression. On the other hand, GW501516-activated PPARδ induced expression of SOCS1, which is a negative regulator of cytokine signal transduction, at the transcriptional level by binding to a PPAR response element in its promoter. This GW501516-mediated induction of SOCS1 expression led to down-regulation of BACE1 expression via inactivation of signal transducer and activator of transcription 1. GW501516-activated PPARδ suppressed the generation of neurotoxic amyloid beta (Aß) in accordance with the decrease in BACE1 expression. Taken together, these results indicate that PPARδ attenuates BACE1 expression via SOCS1-mediated inhibition of signal transducer and activator of transcription 1 signaling, thereby suppressing BACE1-associated generation of neurotoxic Aß.


Assuntos
Secretases da Proteína Precursora do Amiloide/efeitos dos fármacos , Peptídeos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidases/efeitos dos fármacos , Proteína 1 Supressora da Sinalização de Citocina/efeitos dos fármacos , Tiazóis/farmacologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Humanos , Janus Quinase 2/efeitos dos fármacos , Janus Quinase 2/metabolismo , Fator de Transcrição STAT1/metabolismo , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Regulação para Cima
2.
Int J Cancer ; 143(11): 2985-2996, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30204243

RESUMO

Peroxisome proliferator-activated receptor (PPAR) δ is a promising therapeutic target in metabolic and inflammatory disorders. However, its role in oncogenesis is controversial, and its therapeutic potential remains to be determined. In our study, we show that ligand-activated PPARδ forms a complex with the proto-oncogene product c-Myc. The interaction of PPARδ with c-Myc affected the transcriptional activity of c-Myc and the expression of its target genes. The PPARδ-dependent regulation of c-Myc activity was associated with decreased tumorigenicity in breast cancer cells. Administration of the PPARδ ligand GW501516 inhibited tumor growth in xenograft model mice bearing MDA-MB-231 cells stably expressing wild-type PPARδ, but not those expressing dominant-negative PPARδ, by interfering with c-Myc function through protein-protein interaction. Our results indicating that PPARδ forms an antitumorigenic complex with c-Myc in the presence of ligand suggest a potential role of PPARδ in breast cancer development.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Carcinogênese/efeitos dos fármacos , Carcinogênese/metabolismo , PPAR delta/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Tiazóis/farmacologia , Células A549 , Animais , Linhagem Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Ligantes , Células MCF-7 , Células PC12 , Proto-Oncogene Mas , RNA Interferente Pequeno/metabolismo , Ratos
3.
J Cell Biochem ; 119(7): 5609-5619, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29388693

RESUMO

Neuroinflammation-associated release of glutamate from activated microglia has been implicated in the progression of neurodegenerative diseases. However, the regulatory mechanisms underlying this glutamate release are poorly understood. Here, we show that peroxisome proliferator-activated receptor delta (PPARδ) modulates neurotoxicity by inhibiting glutamate release in lipopolysaccharide (LPS)-activated BV-2 microglial cells. Activation of PPARδ by GW501516, a specific PPARδ agonist, inhibited glutamate release in BV-2 cells. This effect of GW501516 was significantly blocked by shRNA-mediated knockdown of PPARδ and by treatment with GSK0660, a specific PPARδ antagonist, indicating that PPARδ is associated with blockade of glutamate release. Additionally, GW501516-activated PPARδ suppressed generation of reactive oxygen species and expression of gp91phox, a functional subunit of NADPH oxidase 2, in BV-2 cells stimulated with LPS. The inhibitory effect of GW501516 on gp91phox expression and glutamate release was further potentiated in the presence of AG490, a specific inhibitor of janus kinase 2 (JAK2), leading to the inhibition of signal transducer and activator of transcription 1 (STAT1). By contrast, GW501516 upregulated the expression of suppressor of cytokine signaling 1 (SOCS1), an endogenous inhibitor of JAK2. Furthermore, neurotoxicity induced by conditioned media from LPS-stimulated BV-2 cells was significantly reduced when conditioned media from BV-2 cells treated with both LPS and GW501516 were used. These results indicate that PPARδ attenuates LPS-triggered neuroinflammation by enhancing SOCS1-mediated inhibition of JAK2/STAT1 signaling, thereby inhibiting neurotoxicity associated with glutamate release.


Assuntos
Ácido Glutâmico/metabolismo , Lipopolissacarídeos/toxicidade , Microglia/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Síndromes Neurotóxicas/tratamento farmacológico , PPAR delta/agonistas , Tiazóis/farmacologia , Animais , Células Cultivadas , Janus Quinase 2/metabolismo , Camundongos , Microglia/metabolismo , Microglia/patologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , PPAR delta/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
4.
Oncotarget ; 8(55): 94091-94103, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-29212212

RESUMO

Migration and invasion of cancer cells into surrounding tissue is a key stage of cancer metastasis. Here, we show that peroxisome proliferator-activated receptor (PPAR) δ regulates migration and invasion of human breast cancer cells via thrombospondin-1 (TSP-1) and its degrading protease, a disintegrin and metalloprotease domains with thrombospondin motifs 1 (ADAMTS1). Activation of PPARδ by GW501516, a specific ligand for PPARδ, led to marked inhibition in the cell migration and TSP-1 expression of breast cancer. These effects were suppressed by small interfering RNA-mediated knock-down of ADAMTS1, indicating that ADAMTS1 is involved in PPARδ-mediated inhibition of migration and TSP-1 expression in breast cancer cells. In addition, ligand-activated PPARδ upregulated expression of ADAMTS1 at the transcriptional level via binding of PPARδ to a direct repeat-1 site within the ADAMTS1 gene promoter. Furthermore, ligand-activated PPARδ suppressed invasion of breast cancer cells in an ADAMTS1-dependent manner. Taken together, these results demonstrate that PPARδ suppresses migration and invasion of breast cancer cells by downregulating TSP-1 in a process mediated by upregulation of ADAMTS1.

5.
Int J Biol Macromol ; 104(Pt A): 281-286, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28602994

RESUMO

Bioactivities of fish collagen peptide are now being elucidated in diverse biological systems. Here, we investigated the effect of fish collagen peptide on the adipogenic differentiation of 3T3-L1 preadipocytes and in obese mice fed a high fat diet (HFD). Subcritical water-hydrolyzed fish collagen peptide (SWFCP) significantly inhibited lipid accumulation during the differentiation of 3T3-L1 preadipocytes, which was accompanied by decreased expression of CCAAT-enhancer-binding protein-α (C/EBP-α), peroxisome proliferator-activated receptor-γ (PPAR-γ), and adipocyte protein 2 (aP2) genes, key regulators of differentiation and maintenance of adipocytes. SWFCP was also found to suppress the palmitate-induced accumulation of lipid vacuoles in hepatocytes. Oral administration of SWFCP significantly reduced HFD-induced body weight gain without a significant difference in food intake. Consistent with its effects in 3T3-L1 preadipocytes, SWFCP inhibited the expression of C/EBP-α, PPAR-γ, and aP2 in epididymal adipose tissue of mice fed a HFD, leading to a significant reduction in adipocyte size. Furthermore, SWFCP significantly reduced serum levels of total cholesterol, triglyceride, and low-density lipoprotein, and increased serum high-density lipoprotein. These observations suggest that SWFCP inhibits adipocyte differentiation through a mechanism involving transcriptional repression of the major adipogenic regulators C/EBP-α and PPAR-γ, thereby reducing body weight gain and adipogenesis in an animal model of obesity.


Assuntos
Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Colágeno/química , Dieta Hiperlipídica/efeitos adversos , Obesidade/tratamento farmacológico , Fragmentos de Peptídeos/farmacologia , Células 3T3-L1 , Adipócitos/citologia , Animais , Peso Corporal/efeitos dos fármacos , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Lipídeos/sangue , Masculino , Camundongos , Obesidade/induzido quimicamente , Obesidade/metabolismo , Obesidade/patologia , PPAR gama/metabolismo , Fragmentos de Peptídeos/uso terapêutico
6.
BMC Complement Altern Med ; 17(1): 212, 2017 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-28403838

RESUMO

BACKGROUND: Dalbergia odorifera T. Chen (Leguminosae) is an indigenous medicinal herb that is widely used as a popular remedy in northern and eastern Asia. However, the cellular mechanisms underlying the biological activity of D. odorifera are not fully elucidated. METHODS: Anti-inflammatory effect of D. odorifera extract (DOE) was determined through intraperitoneal injection in a mouse model of endotoxemia induced by lipopolysaccharide (LPS). RAW 264.7 cells, a murine macrophage, were also treated with LPS to generate a cellular model of inflammation, and investigated the anti-inflammatory activity and underlying mechanisms of DOE and its constituent isoliquiritigenin. RESULTS: DOE dose-dependently inhibited LPS-induced release of high mobility group box 1 (HMGB1), a late proinflammatory cytokine, and decreased cytosolic translocation of HMGB1 in RAW264.7 cells. This inhibitory effect of DOE on HMGB1 release was observed in cells treated with DOE before or after LPS treatment, suggesting that DOE is effective for both treatment and prevention. In addition, DOE significantly inhibited LPS-induced formation of nitric oxide (NO) and expression of inducible NO synthase (iNOS) in a dose-dependent manner. These effects of DOE were accompanied by suppression of HMGB1 release triggered by LPS, suggesting a possible mechanism by which DOE modulates HMGB1 release through NO signaling. Isoriquiritigenin, a constituent of DOE, also attenuated LPS-triggered NO formation and HMGB1 release in RAW264.7 cells, indicating that isoriquiritigenin is an indexing molecule for the anti-inflammatory properties of DOE. Furthermore, c-Jun N-terminal kinase, but not extracellular signal-regulated kinase and p38, mediated DOE-dependent inhibition of HMGB1 release and NO/iNOS induction in RAW 264.7 cells exposed to LPS. Notably, administration of DOE ameliorated survival rates in a mouse model of endotoxemia induced by LPS, where decreased level of circulating HMGB1 was observed. CONCLUSION: These results suggest that DOE confers resistance to LPS-triggered inflammation through NO-mediated inhibitory effects on HMGB1 release.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Dalbergia/química , Endotoxemia/tratamento farmacológico , Proteína HMGB1/antagonistas & inibidores , Fitoterapia , Extratos Vegetais/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/isolamento & purificação , Modelos Animais de Doenças , Endotoxemia/induzido quimicamente , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipopolissacarídeos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico/metabolismo , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos
7.
Pharmacol Res ; 114: 47-55, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27771463

RESUMO

Silent mating type information regulation 2 homolog 1 (SIRT1), a NAD-dependent deacetylase, mediates cellular processes involved in gene silencing and aging. The regulation of lifespan by SIRT1 has been extensively investigated, but less is known about the mechanisms associated with its cellular turnover during inflammatory responses. In this study, we found that peroxisome proliferator-activated receptor (PPAR) γ is associated with SIRT1 stability in murine macrophage RAW 264.7 cells exposed to lipopolysaccharide (LPS). Activation of PPARγ by rosiglitazone, a specific ligand of PPARγ, rescues LPS-induced destabilization of SIRT1, with a concomitant decrease in phosphorylation of residue Ser-46, which is targeted by JNK-1 to promote proteasome-mediated degradation of SIRT1. The rosiglitazone-mediated increase in SIRT1 stability is accompanied by upregulation of mitogen-activated protein kinase phosphatase (MKP)-7, a JNK-specific phosphatase. These effects are significantly influenced by ablation or ectopic expression of PPARγ, indicating that PPARγ is directly involved in the regulation of SIRT1 stability. Furthermore, gain of MKP-7 function mimicked the effect of rosiglitazone on LPS-induced destabilization and ubiquitination of SIRT1. These results indicate that PPARγ-dependent upregulation of MKP-7 improves the stability of SIRT1 by inactivating JNK during inflammatory responses of LPS-activated macrophages.


Assuntos
Fosfatases de Especificidade Dupla/imunologia , Hipoglicemiantes/farmacologia , Lipopolissacarídeos/imunologia , Macrófagos/efeitos dos fármacos , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Fosfatases da Proteína Quinase Ativada por Mitógeno/imunologia , Sirtuína 1/imunologia , Tiazolidinedionas/farmacologia , Animais , Células CHO , Cricetulus , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Proteína Quinase 8 Ativada por Mitógeno/imunologia , PPAR gama/imunologia , Proteólise/efeitos dos fármacos , Células RAW 264.7 , Rosiglitazona , Sirtuína 1/metabolismo , Regulação para Cima/efeitos dos fármacos
8.
Mol Pharmacol ; 90(5): 522-529, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27573670

RESUMO

Peroxisome proliferator-activated receptor δ (PPARδ) has been implicated in vascular pathophysiology. However, its functions in atherogenic changes of the vascular wall have not been fully elucidated. PPARδ activated by GW501516 (2-[2-methyl-4-[[4-methyl-2-[4-(trifluoromethyl)phenyl]-1,3-thiazol-5-yl]methylsulfanyl]phenoxy]acetic acid) significantly inhibited the migration and proliferation of vascular smooth muscle cells (VSMCs) triggered by oxidized low-density lipoprotein (oxLDL). These GW501516-mediated effects were significantly reversed by PPARδ-targeting small-interfering RNA (siRNA), indicating that PPARδ is involved in the action of GW501516. The antiproliferative effect of GW501516 was directly linked to cell cycle arrest at the G0/G1 to S phase transition, which was followed by the down-regulation of cyclin-dependent kinase 4 along with increased levels of p21 and p53. In VSMCs treated with GW501516, the expression of sirtuin 1 (SIRT1) mRNA and protein was time-dependently increased. This GW501516-mediated up-regulation of SIRT1 expression was also demonstrated even in the presence of oxLDL. In addition, GW501516-dependent inhibition of oxLDL-triggered migration and proliferation of VSMCs was almost completely abolished in the presence of SIRT1-targeting siRNA. These effects of GW501516 on oxLDL-triggered phenotypic changes of VSMCs were also demonstrated via activation or inhibition of SIRT1 activity by resveratrol or sirtinol, respectively. Finally, gain or loss of SIRT1 function imitated the action of PPARδ on oxLDL-triggered migration and proliferation of VSMCs. Taken together, these observations indicate that PPARδ-dependent up-regulation of SIRT1 contributes to the antiatherogenic activities of PPARδ by suppressing the migration and proliferation of VSMCs linked to vascular diseases such as restenosis and atherosclerosis.


Assuntos
Movimento Celular/efeitos dos fármacos , Lipoproteínas LDL/farmacologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , PPAR delta/metabolismo , Sirtuína 1/metabolismo , Adenoviridae/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Miócitos de Músculo Liso/efeitos dos fármacos , Interferência de RNA/efeitos dos fármacos , Ratos , Tiazóis
9.
Sci Rep ; 5: 15971, 2015 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-26522327

RESUMO

Inflammatory signal-mediated release of high-mobility group box 1 (HMGB1) is a damage-associated molecular pattern or alarmin. The inflammatory functions of HMGB1 have been extensively investigated; however, less is known about the mechanisms controlling HMGB1 release. We show that SIRT1, the human homolog of the Saccharomyces cerevisiae protein silent information regulator 2, which is involved in cellular senescence and possibly the response to inflammation, forms a stable complex with HMGB1 in murine macrophage RAW264.7 cells. SIRT1 directly interacted with HMGB1 via its N-terminal lysine residues (28-30), and thereby inhibited HMGB1 release to improve survival in an experimental model of sepsis. By contrast, inflammatory stimuli such as lipopolysaccharide (LPS) and tumor necrosis factor-α promoted HMGB1 release by provoking its dissociation from SIRT1 dependent on acetylation, thereby increasing the association between HMGB1 and chromosome region maintenance 1, leading to HMGB1 translocation. In vivo infection with wild-type SIRT1 and HMGB1(K282930R), a hypo-acetylation mutant, improved survival (85.7%) during endotoxemia more than infection with wild-type SIRT1 and HMGB1-expressing adenovirus, indicating that the acetylation-dependent interaction between HMGB1 and SIRT1 is critical for LPS-induced lethality. Taken together, we propose that SIRT1 forms an anti-inflammatory complex with HMGB1, allowing cells to bypass the response to inflammation.


Assuntos
Endotoxemia/metabolismo , Proteína HMGB1/metabolismo , Sirtuína 1/metabolismo , Acetilação , Animais , Linhagem Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células HEK293 , Células HL-60 , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Camundongos , Sepse/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Células U937
10.
Am J Cancer Res ; 4(6): 674-82, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25520859

RESUMO

Peroxisome proliferator-activated receptor (PPAR) δ is implicated in the carcinogenesis of several types of cancer. However, the therapeutic efficacy of PPARδ ligands against cancer progression is unclear. Here, we showed that PPARδ modulates the migration and invasion of melanoma cells by up-regulating Snail expression. Activation of PPARδ by GW501516, a specific ligand for PPARδ, significantly increased the migration and invasion of highly metastatic A375SM cells, but not that of low metastatic A375P cells. The migration- and invasion-promoting effects of PPARδ on A375SM cells was associated with increased Snail expression, which was accompanied by a decrease in E-cadherin expression. Furthermore, a significant concentration- and time-dependent increase in the levels of Snail mRNA and protein was observed in A375SM cells (but not A375P cells) treated with GW501516. The effects of GW501516 were almost completely abrogated by a small interfering RNA against PPARδ, suggesting that PPARδ mediates the effects of GW501516. Activation of PPARδ in SK-MEL-2 and SK-MEL-5 (but not SK-MEL-3) melanoma cell lines also led to significant increases in the expression of Snail mRNA and protein, which mirrored the invasive and migratory potential of these cell lines. These results suggest that PPARδ promotes the aggressive phenotype observed in highly metastatic melanoma cells by up-regulating Snail.

11.
J Vasc Res ; 51(3): 221-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25116733

RESUMO

We investigated the role of peroxisome proliferator-activated receptor (PPAR) δ on angiotensin (Ang) II-induced activation of matrix metalloproteinase (MMP)-2 in vascular smooth muscle cells (VSMCs). Activation of PPARδ by GW501516, a specific ligand for PPARδ, attenuated Ang II-induced activation of MMP-2 in a concentration-dependent manner. GW501516 also inhibited the generation of reactive oxygen species in VSMCs treated with Ang II. A marked increase in the mRNA levels of tissue inhibitor of metalloproteinase (TIMP)-2 and -3, endogenous antagonists of MMPs, was also observed in GW501516-treated VSMCs. These effects were markedly reduced in the presence of siRNAs against PPARδ, indicating that the effects of GW501516 are PPARδ dependent. Among the protein kinases inhibited by GW501516, suppression of phosphatidylinositol 3-kinase/Akt signaling was shown to have the greatest effect on activation of MMP-2 in VSMCs treated with Ang II. Concomitantly, GW501516-mediated inhibition of MMP-2 activation in VSMCs treated with Ang II was associated with the suppression of cell migration to levels approaching those in cells not exposed to Ang II. Thus, activation of PPARδ confers resistance to Ang II-induced degradation of the extracellular matrix by upregulating expression of its endogenous inhibitor TIMP and thereby modulating cellular responses to Ang II in vascular cells.


Assuntos
Metaloproteinase 2 da Matriz/metabolismo , Miócitos de Músculo Liso/metabolismo , PPAR delta/metabolismo , Angiotensina II/farmacologia , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Masculino , Músculo Liso Vascular/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Ratos , Transdução de Sinais/efeitos dos fármacos , Tiazóis
12.
J Dermatol Sci ; 76(1): 44-50, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25149191

RESUMO

BACKGROUND: Changes in skin connective tissues mediated by ultraviolet (UV) radiation have been suggested to cause the skin wrinkling normally associated with premature aging of the skin. Recent investigations have shown that peroxisome proliferator-activated receptor (PPAR) δ plays multiple biological roles in skin homeostasis. OBJECTIVE: We attempted to investigate whether PPARδ modulates elastin protein levels and secretion of matrix metalloproteinase (MMP)-2 in UVB-irradiated human dermal fibroblasts (HDFs) and mouse skin. METHODS: These studies were undertaken in primary HDFs or HR-1 hairless mice using Western blot analyses, small interfering (si)RNA-mediated gene silencing, and Fluorescence microscopy. RESULTS: In HDFs, UVB irradiation induced increased secretion of MMP-2 and reduced levels of elastin. Activation of PPARδ by GW501516, a ligand specific for PPARδ, markedly attenuated UVB-induced MMP-2 secretion with a concomitant increase in the level of elastin. These effects were reduced by the presence of siRNAs against PPARδ or treatment with GSK0660, a specific inhibitor of PPARδ. Furthermore, GW501516 elicited a dose- and time-dependent increase in the expression of elastin. Modulation of MMP-2 secretion and elastin levels by GW501516 was associated with a reduction in reactive oxygen species (ROS) production in HDFs exposed to UVB. Finally, in HR-1 hairless mice, administration of GW501516 significantly reduced UVB-induced MMP-2 expression with a concomitant increase in elastin levels, and these effects were significantly reduced by the presence of GSK0660. CONCLUSION: Our results suggest that PPARδ-mediated modulation of MMP-2 secretion and elastin expression may contribute to the maintenance of skin integrity by inhibiting ROS generation.


Assuntos
Elastina/metabolismo , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Metaloproteinase 2 da Matriz/metabolismo , PPAR delta/metabolismo , Pele/metabolismo , Animais , Relação Dose-Resposta à Radiação , Inativação Gênica , Homeostase , Humanos , Camundongos , Microscopia de Fluorescência , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sulfonas/química , Tiofenos/química , Raios Ultravioleta , Cicatrização
13.
Pharm Biol ; 50(10): 1281-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22857151

RESUMO

CONTEXT: Alisma orientale (Sam.) Juzepczuk (Alismataceae) is an indigenous medicinal herb that has been traditionally used for diuretic, hypolipidemic, anti-inflammatory, and antidiabetic proposes in northern and eastern Asia. OBJECTIVE: This study examined the mechanisms underlying the cytoprotective effect of an aqueous extract of A. orientale (AEAO) against long-chain saturated fatty acid-induced cellular injury. MATERIALS AND METHODS: HepG2 cells were treated with 0.5 mM palmitate to generate a cellular model of nonalcoholic fatty liver disease (NAFLD). Using this cellular model, the cytoprotective effect of AEAO (100 µg/mL) against long-chain saturated fatty acid-induced cellular injury was evaluated by measuring the steatosis, ROS accumulation, and apoptosis. RESULTS: AEAO significantly attenuated palmitate-induced intracellular steatosis and cellular damage up to 54 and 33%, respectively. Palmitate-induced intracellular levels of reactive oxygen species (ROS) and reactive aldehydes were significantly reduced in the presence of AEAO to 40 and 75%, respectively, suggesting that oxidative stress plays a role in the palmitate-induced damage. AEAO inhibited the palmitate-mediated activation of c-Jun NH(2)-terminal kinase (JNK), a kinase that is correlated with NAFLD. Inhibition of JNK by SP600125 or addition of AEAO significantly reduced palmitate-induced steatosis, ROS accumulation, and apoptosis, indicating that the protective effects of AEAO against palmitate-induced cellular damage result from blocking ROS-activated JNK signaling. DISCUSSION AND CONCLUSION: The combined properties of AEAO in cellular steatosis and ROS production are beneficial for treating NAFLD, which includes complex metabolic changes, such that modulation of a single target is often not sufficient to achieve the desired therapeutic effect.


Assuntos
Alisma/química , Antioxidantes/farmacologia , Fígado Gorduroso/patologia , Extratos Vegetais/farmacologia , Antioxidantes/isolamento & purificação , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular , Citoproteção/efeitos dos fármacos , Fígado Gorduroso/tratamento farmacológico , Células Hep G2 , Hepatoblastoma/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Medicina Tradicional do Leste Asiático , Hepatopatia Gordurosa não Alcoólica , Estresse Oxidativo/efeitos dos fármacos , Palmitatos/toxicidade , Espécies Reativas de Oxigênio/metabolismo
14.
Free Radic Res ; 46(7): 912-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22519881

RESUMO

Angiotensin II (Ang II)-mediated modification of the redox milieu of vascular smooth muscle cells (VSMCs) has been implicated in several pathophysiological processes, including cell proliferation, migration and differentiation. In this study, we demonstrate that the peroxisome proliferator-activated receptor (PPAR) δ counteracts Ang II-induced production of reactive oxygen species (ROS) in VSMCs. Activation of PPARδ by GW501516, a specific ligand for PPARδ, significantly reduced Ang II-induced ROS generation in VSMCs. This effect was, however, reversed in the presence of small interfering (si)RNA against PPARδ. The marked increase in ROS levels induced by Ang II was also eliminated by the inhibition of phosphatidylinositol 3-kinase (PI3K) but not of protein kinase C, suggesting the involvement of the PI3K/Akt signalling pathway in this process. Accordingly, ablation of Akt with siRNA further enhanced the inhibitory effects of GW501516 in Ang II-induced superoxide production. Ligand-activated PPARδ also blocked Ang II-induced translocation of Rac1 to the cell membrane, inhibiting the activation of NADPH oxidases and consequently ROS generation. These results indicate that ligand-activated PPARδ plays an important role in the cellular response to oxidative stress by decreasing ROS generated by Ang II in vascular cells.


Assuntos
Angiotensina II/farmacologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , PPAR gama/agonistas , Espécies Reativas de Oxigênio/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Angiotensina II/antagonistas & inibidores , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , PPAR gama/genética , PPAR gama/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo
15.
Biochem J ; 444(1): 27-38, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22335598

RESUMO

UV radiation-mediated photodamage to the skin has been implicated in premature aging and photoaging-related skin cancer and melanoma. Little is known about the cellular events that underlie premature senescence, or how to impede these events. In the present study we demonstrate that PPARδ (peroxisome-proliferator-activated receptor δ) regulates UVB-induced premature senescence of normal keratinocytes. Activation of PPARδ by GW501516, a specific ligand of PPARδ, significantly attenuated UVB-mediated generation of ROS (reactive oxygen species) and suppressed senescence of human keratinocytes. Ligand-activated PPARδ up-regulated the expression of PTEN (phosphatase and tensin homologue deleted on chromosome 10) and suppressed the PI3K (phosphatidylinositol 3-kinase)/Akt pathway. Concomitantly, translocation of Rac1 to the plasma membrane, which leads to the activation of NADPH oxidases and generation of ROS, was significantly attenuated. siRNA (small interfering RNA)-mediated knockdown of PTEN abrogated the effects of PPARδ on cellular senescence, on PI3K/Akt/Rac1 signalling and on generation of ROS in keratinocytes exposed to UVB. Finally, when HR-1 hairless mice were treated with GW501516 before exposure to UVB, the number of senescent cells in the skin was significantly reduced. Thus ligand-activated PPARδ confers resistance to UVB-induced cellular senescence by up-regulating PTEN and thereby modulating PI3K/Akt/Rac1 signalling to reduce ROS generation in keratinocytes.


Assuntos
Queratinócitos/efeitos da radiação , PTEN Fosfo-Hidrolase/fisiologia , Superóxidos/antagonistas & inibidores , Animais , Membrana Celular/metabolismo , Células Cultivadas , Senescência Celular , Criança , Ativação Enzimática , Humanos , Queratinócitos/fisiologia , Ligantes , Camundongos , Camundongos Pelados , PPAR delta/agonistas , PPAR delta/fisiologia , PTEN Fosfo-Hidrolase/biossíntese , Fosfatidilinositol 3-Quinases/fisiologia , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Pele/citologia , Pele/metabolismo , Pele/efeitos da radiação , Superóxidos/metabolismo , Tiazóis/farmacologia , Raios Ultravioleta , Regulação para Cima , Proteínas rac1 de Ligação ao GTP/metabolismo
16.
J Biol Chem ; 286(52): 44585-93, 2011 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-22072715

RESUMO

Cellular senescence-associated changes in blood vessels have been implicated in aging and age-related cardiovascular disorders. Here, we demonstrate that peroxisome proliferator-activated receptor (PPAR) δ coordinates angiotensin (Ang) II-induced senescence of human vascular smooth muscle cells (VSMCs). Activation of PPARδ by GW501516, a specific ligand for PPARδ, significantly attenuated Ang II-induced generation of superoxides and suppressed senescence of VSMCs. A marked increase in the levels of p53 and p21 induced by Ang II was blunted by the treatment with GW501516. Ligand-activated PPARδ up-regulated expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and suppressed the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Knockdown of PTEN with siRNA abrogated the effects of PPARδ on cellular senescence, on PI3K/Akt signaling, and on generation of ROS in VSMCs treated with Ang II. Finally, administration of GW501516 to apoE-deficient mice treated with Ang II significantly reduced the number of senescent cells in the aorta, where up-regulation of PTEN with reduced levels of phosphorylated Akt and ROS was demonstrated. Thus, ligand-activated PPARδ confers resistance to Ang II-induced senescence by up-regulation of PTEN and ensuing modulation of the PI3K/Akt signaling to reduce ROS generation in vascular cells.


Assuntos
Angiotensina II/metabolismo , Senescência Celular/fisiologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Superóxidos/metabolismo , Substituição de Aminoácidos , Angiotensina II/genética , Animais , Aorta/citologia , Aorta/metabolismo , Células Cultivadas , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Mutantes , Músculo Liso Vascular/citologia , Mutação de Sentido Incorreto , Miócitos de Músculo Liso/citologia , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores Citoplasmáticos e Nucleares/genética , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
17.
Free Radic Biol Med ; 50(6): 680-8, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21182935

RESUMO

Although aldose reductase (AR) has been implicated in the cellular response to oxidative stress, the role of AR in ultraviolet-B (UVB)-induced cellular injury has not been investigated. Here, we show that an increased expression of AR in human keratinocytes modulates UVB-induced apoptotic cell death and senescence. Overexpression of AR in HaCaT cells significantly attenuated UVB-induced cellular damage and apoptosis, with a decreased generation of reactive oxygen species (ROS) and aldehydes. Ablation of AR with small interfering RNA or inhibition of AR activity abolished these effects. We also show that increased AR activity suppressed UVB-induced activation of the p38 and c-Jun N-terminal kinases, but did not affect the extracellular signal-regulated kinase and phosphatidylinositol 3-kinase pathways. Similarly, UVB-induced translocation of Bax and Bcl-2 to mitochondria and cytosol, respectively, was markedly attenuated in cells overexpressing AR. Knockdown or inhibition of AR activity in primary cultured keratinocytes enhanced UVB-induced cellular senescence and increased the level of a cell-cycle regulatory protein, p53. Finally, cellular apoptosis induced by UVB radiation was significantly reduced in the epidermis of transgenic mice overexpressing human AR. These findings suggest that AR plays an important role in the cellular response to oxidative stress by sequestering ROS and reactive aldehydes generated in keratinocytes.


Assuntos
Aldeído Redutase/metabolismo , Apoptose , Senescência Celular , Queratinócitos/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Aldeído Redutase/genética , Aldeídos/metabolismo , Animais , Células Cultivadas , Epiderme/metabolismo , Fibroblastos/metabolismo , Expressão Gênica , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Fosfatidilinositol 3-Quinase/genética , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Queimadura Solar/metabolismo , Queimadura Solar/patologia , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Proteína X Associada a bcl-2/metabolismo
18.
Free Radic Res ; 42(11-12): 930-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19031314

RESUMO

Aldose reductase (AR) is abundantly expressed in a variety of cell lineages and has been implicated in the cellular response against oxidative stress. However, the exact functional role of AR against oxidative stress remains relatively unclear. This study investigated the role of AR in acrolein- or hydrogen peroxide-induced apoptosis using the J774.A.1 macrophage cell line. Ablation of AR with a small interference RNA or inhibition of AR activity significantly enhanced the acrolein- or hydrogen peroxide-induced generation of reactive oxygen species and aldehydes, leading to increased apoptotic cell death. Blockade of AR activity in J774A.1 cells markedly augmented the acrolein- or hydrogen peroxide-induced translocation of Bax to mitochondria along with reduced Bcl-2 and increased release of cytochrome c from the mitochodria. Taken together, these findings indicate that AR plays an important role in the cellular response against oxidative stress, by sequestering the reactive molecules generated in cells exposed to toxic substances.


Assuntos
Acroleína/farmacologia , Aldeído Redutase/metabolismo , Regulação para Baixo , Peróxido de Hidrogênio/farmacologia , Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/biossíntese , Aldeído Redutase/genética , Aldeídos/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocromos c/metabolismo , Regulação para Baixo/efeitos dos fármacos , Glucose Oxidase/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Proteína X Associada a bcl-2/metabolismo
19.
Apoptosis ; 13(10): 1223-31, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18690538

RESUMO

Yeast-based functional screening of a human glioblastoma cDNA library identified ras-related nuclear protein (Ran) as a novel suppressor of Bcl-2-associated X protein (Bax), a pro-apoptotic member of the Bcl-2 family of proteins. Yeast cells that expressed human Ran were resistant to Bax-induced cell death. In U373MG glioblastoma cells, stable overexpression of Ran significantly attenuated apoptotic cell death induced by the chemotherapeutic agent paclitaxel. FACS analysis demonstrated that Ran is involved in paclitaxel-induced cell cycle arrest. Stable overexpression of Ran also markedly inhibited the phosphorylation of Bcl-2 by paclitaxel, and inhibited the translocation of Bax, the release of cytochrome c and activation of caspase-3. Paclitaxel-induced phosphorylation of c-JUN N-terminal kinase (JNK), but not p38, extracellular signal-regulated kinase and Akt, was markedly suppressed in U373MG cells that stably expressed Ran. These results suggest that Ran suppresses paclitaxel-induced cell death through the downregulation of JNK-mediated signal pathways.


Assuntos
Apoptose/efeitos dos fármacos , Glioblastoma/patologia , Paclitaxel/farmacologia , Proteína ran de Ligação ao GTP/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Biblioteca Gênica , Glioblastoma/enzimologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
20.
Pharmacol Res ; 58(1): 15-21, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18588981

RESUMO

The osmotic response element (ORE) differs from the nuclear factor-kappaB (NF-kappaB) binding sequence by a single base pair; therefore, we investigated the involvement of NF-kappaB in the induction of aldose reductase (AR) by curcumin. Curcumin, an herb-derived polyphenolic compound, elicited an increase in the expression and promoter activity of the AR gene in a nuclear factor-erythroid 2-related factor 2 (Nrf2)-dependent manner. Small interfering RNA (siRNA) against p65 or BAY11-7082, an inhibitor of NF-kappaB, significantly suppressed the curcumin and/or Nrf2-induced increase in expression levels and promoter activity of the AR gene. BAY11-7082 or siRNA against p65 also attenuated the curcumin-induced increase in the promoter activity of the wild type AR-ORE(wt) gene, but not that of the mutated AR-ORE(mt), indicating that the ORE is essential for the response to NF-kappaB. The expression of p65, the promoter activity and DNA binding activity of NF-kappaB were enhanced in the presence of curcumin in cells that were transfected with Nrf2 compared to those treated with curcumin alone. Cells that had been preincubated with curcumin demonstrated resistance to reactive oxygen species-induced cell damage through the suppressive effects in the generation of reactive aldehydes. These effects were significantly attenuated in the presence of BAY11-7082, indicating the involvement of NF-kappaB in the cellular response of AR to oxidative stress and toxic aldehydes.


Assuntos
Aldeído Redutase/biossíntese , Curcumina/farmacologia , Fator 2 Relacionado a NF-E2/fisiologia , NF-kappa B/fisiologia , Nitrilas/farmacologia , Sulfonas/farmacologia , Aldeído Redutase/genética , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação Enzimológica da Expressão Gênica , Técnicas In Vitro , Masculino , NF-kappa B/antagonistas & inibidores , Subunidade p50 de NF-kappa B/genética , Osmose , Estresse Oxidativo , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Elementos de Resposta , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA