Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 297
Filtrar
1.
Trends Pharmacol Sci ; 45(1): 9-23, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38065777

RESUMO

Peroxisome proliferator-activated receptors [PPARs; PPARα, PPARß/δ (also known as PPARδ), and PPARγ] widely recognized for their important role in glucose/lipid homeostasis, have recently received significant attention due to their additional anti-inflammatory and neuroprotective effects. Several newly developed PPAR agonists have shown high selectivity for specific PPAR isoforms in vitro and in vivo, offering the potential to achieve desired therapeutic outcomes while reducing the risk of adverse effects. In this review, we discuss the latest preclinical and clinical studies of the activation of PPARs by synthetic, natural, and isoform-specific (full, partial, and dual) agonists for the treatment of neuroinflammatory diseases, including HIV-associated neurocognitive disorders (HAND), Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and cerebral ischemia.


Assuntos
PPAR delta , PPAR beta , Humanos , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Doenças Neuroinflamatórias , PPAR delta/agonistas , PPAR delta/fisiologia , PPAR beta/fisiologia , PPAR alfa/agonistas , PPAR alfa/fisiologia , PPAR gama/agonistas , PPAR gama/fisiologia , Hipoglicemiantes
2.
Life Sci ; 291: 120239, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-34942163

RESUMO

Aim Investigate whether inheritance of improved skeletal muscle mitochondrial function and its association with glycemic control are multigenerational benefits of exercise. MAIN METHODS: Male Swiss mice were subjected to 8 weeks of endurance training and mated with untrained females. KEY FINDINGS: Trained fathers displayed typical endurance training-induced adaptations. Remarkably, offspring from trained fathers also exhibited higher endurance performance, mitochondrial oxygen consumption, glucose tolerance and insulin sensitivity. However, PGC-1α expression was not increased in the offspring. In the offspring, the expression of the co-repressor NCoR1 was reduced, increasing activation of PGC-1α target genes. These effects correlated with higher DNA methylation at the NCoR1 promoter in both, the sperm of trained fathers and in the skeletal muscle of their offspring. SIGNIFICANCE: Higher skeletal muscle mitochondrial function is inherited by epigenetic de-activation of a key PGC-1α co-repressor.


Assuntos
Mitocôndrias/metabolismo , Condicionamento Físico Animal/fisiologia , Esforço Físico/fisiologia , Animais , Metilação de DNA , Epigênese Genética/genética , Feminino , Masculino , Camundongos , Mitocôndrias/fisiologia , Músculo Esquelético/fisiologia , Correpressor 1 de Receptor Nuclear/metabolismo , Consumo de Oxigênio/fisiologia , Herança Paterna/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Condicionamento Físico Animal/métodos , RNA Mensageiro/genética
3.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34445679

RESUMO

It has been more than three decades since peroxisome proliferator-activated receptors (PPARs) were first discovered. Many investigations have revealed the central regulators of PPARs in lipid and glucose homeostasis in response to different nutrient conditions. PPARs have attracted much attention due to their ability to improve metabolic syndromes, and they have also been proposed as classical drug targets for the treatment of hyperlipidemia and type 2 diabetes (T2D) mellitus. In parallel, adipose tissue is known to play a unique role in the pathogenesis of insulin resistance and metabolic syndromes due to its ability to "safely" store lipids and secrete cytokines that regulate whole-body metabolism. Adipose tissue relies on a complex and subtle network of transcription factors to maintain its normal physiological function, by coordinating various molecular events, among which PPARs play distinctive and indispensable roles in adipocyte differentiation, lipid metabolism, adipokine secretion, and insulin sensitivity. In this review, we discuss the characteristics of PPARs with special emphasis on the roles of the different isotypes in adipocyte biology.


Assuntos
Tecido Adiposo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Adipócitos/fisiologia , Homeostase , Resistência à Insulina/fisiologia , Metabolismo dos Lipídeos/fisiologia
4.
Exp Eye Res ; 208: 108617, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34010603

RESUMO

Peroxisome Proliferator-Activated Receptors (PPARs) are a family of nuclear receptors that play essential roles in modulating cell differentiation, inflammation, and metabolism. Three subtypes of PPARs are known: PPAR-alpha (PPARα), PPAR-gamma (PPARγ), and PPAR-beta/delta (PPARß/δ). PPARα activation reduces lipid levels and regulates energy homeostasis, activation of PPARγ results in regulation of adipogenesis, and PPARß/δ activation increases fatty acid metabolism and lipolysis. PPARs are linked to various diseases, including but not limited to diabetes, non-alcoholic fatty liver disease, glaucoma and atherosclerosis. In the past decade, numerous studies have assessed the functional properties of PPARs in the eye and key PPAR mechanisms have been discovered, particularly regarding the retina and cornea. PPARγ and PPARα are well established in their functions in ocular homeostasis regarding neuroprotection, neovascularization, and inflammation, whereas PPARß/δ isoform function remains understudied. Naturally, studies on PPAR agonists and antagonists, associated with ocular pathology, have also gained traction with the development of PPAR synthetic ligands. Studies on PPARs has significantly influenced novel therapeutics for diabetic eye disease, ocular neuropathy, dry eye, and age-related macular degeneration (AMD). In this review, therapeutic potentials and implications will be highlighted, as well as reported adverse effects. Further investigations are necessary before any of the PPARs ligands can be utilized, in the clinics, to treat eye diseases. Future research on the prominent role of PPARs will help unravel the complex mechanisms involved in order to prevent and treat ocular diseases.


Assuntos
Oftalmopatias/metabolismo , Metabolismo dos Lipídeos/fisiologia , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Animais , Homeostase , Humanos , Ligantes
5.
Shock ; 56(6): 1066-1079, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33988537

RESUMO

OBJECTIVES: To explore the role of LPS binding protein (LBP) in metabolism and optimize sepsis treatment. DESIGN: A sepsis model was established by injecting LPS into LBP-/- rats and WT rats and observing changes in the liver over time (0, 1, 6, and 24 h). SETTING: Detecting liver inflammation and injury. Optimizing the treatment of sepsis. SUBJECTS: WT rats and LBP-/- rats. INTERVENTIONS: We established a sepsis model by injecting LPS intravenously. MEASUREMENTS AND MAIN RESULTS: First, we induced sepsis in WT and LBP-/- rats with LPS. The rats were sacrificed, and serum and liver samples were collected at 1, 6, and 24 h after LPS injection. We found that the deletion of LBP reduced LPS-induced liver inflammation and injury at 1 and 6 h. Ballooning degeneration was clearly present in LBP-/- rat livers at 24 h after LPS injection. We found that mitochondrial damage and reactive oxygen species (ROS) levels were higher in LBP-/- rat livers than in WT rat livers at 24 h after LPS injection. According to the transcriptomic results, the peroxisome proliferator-activated receptor (PPAR) pathway may be the reason for lesions in LBP-/- rats. To further investigate the function of PPARα in sepsis, we inhibited mTOR with rapamycin and examined mitochondrial injury and ROS levels. The levels of mitochondrial damage and ROS were reduced after LBP-/- rats were pretreated with rapamycin in the context of LPS-induced sepsis. Inhibiting CYP4a2, one of the PPARα-target gene products, reduced the level of LPS-induced ROS in LBP-/- rats. CONCLUSION: LBP protects hepatic mitochondria against LPS-induced damage via the LBP-PPARα-CYP4a2 signaling pathway.


Assuntos
Proteínas de Fase Aguda/fisiologia , Proteínas de Transporte/fisiologia , Sistema Enzimático do Citocromo P-450/fisiologia , Glicoproteínas de Membrana/fisiologia , Mitocôndrias Hepáticas/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Sepse/metabolismo , Transdução de Sinais/fisiologia , Animais , Modelos Animais de Doenças , Ratos
6.
Circ Res ; 128(7): 1021-1039, 2021 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-33793338

RESUMO

Nuclear receptors represent a large family of ligand-activated transcription factors which sense the physiological environment and make long-term adaptations by mediating changes in gene expression. In this review, we will first discuss the fundamental mechanisms by which nuclear receptors mediate their transcriptional responses. We will focus on the PPAR (peroxisome proliferator-activated receptor) family of adopted orphan receptors paying special attention to PPARγ, the isoform with the most compelling evidence as an important regulator of arterial blood pressure. We will review genetic data showing that rare mutations in PPARγ cause severe hypertension and clinical trial data which show that PPARγ activators have beneficial effects on blood pressure. We will detail the tissue- and cell-specific molecular mechanisms by which PPARs in the brain, kidney, vasculature, and immune system modulate blood pressure and related phenotypes, such as endothelial function. Finally, we will discuss the role of placental PPARs in preeclampsia, a life threatening form of hypertension during pregnancy. We will close with a viewpoint on future research directions and implications for developing novel therapies.


Assuntos
Pressão Sanguínea/fisiologia , Hipertensão/genética , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Animais , Encéfalo/metabolismo , Feminino , Humanos , Sistema Imunitário/fisiologia , Rim/metabolismo , Camundongos , PPAR gama/genética , PPAR gama/fisiologia , Receptores Ativados por Proliferador de Peroxissomo/genética , Placenta , Pré-Eclâmpsia/etiologia , Gravidez , Ratos , Pesquisa , Fatores de Transcrição/fisiologia
7.
Int J Mol Sci ; 22(3)2021 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-33540894

RESUMO

Heart failure remains the most common cause of death in the industrialized world. In spite of new therapeutic interventions that are constantly being developed, it is still not possible to completely protect against heart failure development and progression. This shows how much more research is necessary to understand the underlying mechanisms of this process. In this review, we give a detailed overview of the contribution of impaired mitochondrial dynamics and energy homeostasis during heart failure progression. In particular, we focus on the regulation of fatty acid metabolism and the effects of fatty acid accumulation on mitochondrial structural and functional homeostasis.


Assuntos
Ácidos Graxos/metabolismo , Insuficiência Cardíaca/metabolismo , Mitocôndrias Cardíacas/metabolismo , Dinâmica Mitocondrial , Tecido Adiposo/metabolismo , Sinalização do Cálcio , Cardiomiopatias/metabolismo , Ceramidas/biossíntese , Ciclo do Ácido Cítrico , Progressão da Doença , Ácidos Graxos/efeitos adversos , Homeostase , Humanos , Corpos Cetônicos/metabolismo , Doenças Mitocondriais/metabolismo , Mitofagia , NAD/metabolismo , Pericárdio/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Espécies Reativas de Oxigênio/metabolismo
8.
Biomed Pharmacother ; 127: 110127, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32325349

RESUMO

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a commonly-encountered chronic liver disease which lacks verified pharmacological interventions. Gan-Jiang-Ling-Zhu decoction (GJLZ) is a classic formula utilized in clinical practice. In this study, we aimed to evaluate the therapeutic effect of GJLZ in NAFLD and explore the possible underlying mechanisms. METHODS: Twenty-four rats were randomly divided into three groups: normal group, fed with chow diet for 8 weeks; model group, fed with high fat diet for 8 weeks; and GJLZ group, initially fed HFD for 4 weeks, and then administered the GJLZ decoction for 4 weeks by oral gavage while continuously feeding HFD. Rats were sacrificed after the intervention, and liver tissues and blood samples were harvested. Liver steatosis was detected by HE and Oil Red O staining. Body weight and liver index were analyzed. Liver triglyceride (TG), total cholesterol (TC), and low-density lipoprotein (LDL), serum almandine aminotransferase (ALT), aspartate aminotransferase (AST), and nonesterified fatty acid (NEFA) were assayed using commercial kits. Differentially expressed genes were identified by RNA-sequencing and verified using real-time PCR (RT-PCR) and western blotting. Whole miRNAs were detected by RNA-sequence analysis, and mRNA-targeted miRNAs were verified by RT-PCR. The miRNA-mRNA regulation pattern was confirmed using the dual-luciferase reporter assay. RESULTS: Treatment with GJLZ significantly improved hepatic steatosis and inflammation, reduced liver index and liver TG content, and also significantly reduced serum ALT and AST levels. Based on the results of RNA-sequence analysis, five differentially expressed genes (DEGs) in the peroxisome proliferator-activated receptor (PPAR) signaling pathway were recognized. RT-PCR confirmed that carnitine palmitoyltransferase 1b (CPT1B) expression was significantly regulated by GJLZ treatment. GJLZ decoction intervention also increased significantly hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA) expression. Next, miRNA profiling and screening were performed based on CPT1B alteration. Rno-miR-138-5p likely responded to GJLZ intervention, and rno-miR-138-5p inhibitor increased CPT1B expression while rno-miR-138-5p mimic reduced CPT1B expression. When CPT1B mutated, miR-138-5p mimic and inhibitor could not regulate the luciferase activity of CPT1B. CONCLUSIONS: GJLZ is an effective formula for NAFLD management, and its possible mechanism of action involves the regulation of CPT1B expression via rno-miR-138-5p.


Assuntos
Carnitina O-Palmitoiltransferase/genética , Medicamentos de Ervas Chinesas/uso terapêutico , Medicina Tradicional Chinesa , MicroRNAs/fisiologia , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Animais , Carnitina O-Palmitoiltransferase/fisiologia , Medicamentos de Ervas Chinesas/farmacologia , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/metabolismo , Masculino , Hepatopatia Gordurosa não Alcoólica/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Ratos , Ratos Wistar , Triglicerídeos/metabolismo
9.
Domest Anim Endocrinol ; 67: 42-53, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30690257

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are members of a nuclear receptor family of ligand-dependent transcription factors. Three isoforms of PPAR named PPARα, PPARß/δ, and PPARγ have been described, each encoded by a separate gene: PPARA, PPARD, and PPARG, respectively. In the present study, we examined the profiles of PPAR and retinoid X receptor (RXR; PPAR heterodimer partner) mRNA expression and PPAR DNA binding activity in porcine trophoblast tissue collected on days 15, 20, 25, and 30 of pregnancy and in day-20 embryos. Placenta trophoblast cells isolated on day 25 of pregnancy were used to determine effects of (1) cytokines on PPAR and RXR mRNA expression and (2) PPAR agonists on prostaglandin (PG) E2 synthesis and the expression of genes involved in steroidogenesis, fatty acid binding, and PG transport, as well as on cell proliferation. The mRNA expression of PPARA and RXRB was greater in trophoblast tissue collected on days 25 and 30 of pregnancy compared with day 15 (P < 0.05), while DNA binding activity of PPARα decreased between day 15 and 25 (P < 0.05). Increased concentrations of PPARD and RXRA transcripts were observed in trophoblasts collected on day 20 compared to trophoblasts from days 15 and 30 (P < 0.05). Moreover, concentrations of DNA-bound PPARß/δ and PPARγ proteins increased in day-30 trophoblasts compared to day 15 (P < 0.01) and day 20 (P < 0.05), respectively. On day 20 of gestation, the mRNA expression of PPARD, PPARG, and RXRA and protein levels of PPARα and PPARγ isoforms were greater in trophoblast than embryonic tissue (P < 0.01). Interleukin 1ß and/or interferon γ, but not IL6 and leukemia inhibitory factor, upregulated PPAR and RXR mRNA expression in placenta trophoblast cells in vitro (P < 0.05). Rosiglitazone (a PPARγ agonist) stimulated prostaglandin E synthase mRNA expression in trophoblast cells and PGE2 accumulation in incubation medium (P < 0.05). Moreover, activation of PPAR isoforms differentially affected the expression of genes involved in steroidogenesis, fatty acid binding, and PG transport in studied cells. Finally, PPARα and PPARγ agonists stimulated trophoblast cell proliferation (P < 0.05), and this effect was abolished by the addition of a respective PPAR antagonist (P < 0.05). Overall, these results point to a role of PPAR isoforms in porcine placenta development and function.


Assuntos
Receptores Ativados por Proliferador de Peroxissomo/genética , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Sus scrofa/embriologia , Trofoblastos/química , Trofoblastos/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Citocinas/farmacologia , DNA/metabolismo , Dinoprostona/biossíntese , Implantação do Embrião/fisiologia , Embrião de Mamíferos/química , Embrião de Mamíferos/fisiologia , Feminino , Expressão Gênica/efeitos dos fármacos , Placenta , Placentação/fisiologia , Gravidez , RNA Mensageiro/análise , Receptores X de Retinoides/genética , Sus scrofa/fisiologia , Trofoblastos/citologia
10.
Theriogenology ; 127: 88-101, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30677596

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor family of ligand-dependent transcription factors. PPARs are important regulators of glucose and fatty acid metabolism, apoptosis, angiogenesis, cell proliferation and differentiation, and immune response. Their possible role in the female reproductive tract was demonstrated. In the present study, cultured luminal epithelial (LE) and stromal (ST) cells of the porcine endometrium were used to examine (1) the effect of conceptus exposed medium (CEM) on mRNA and protein expression and DNA binding activity of PPARA, PPARD, and PPARG isoforms, and (2) the effect of PPARA, PPARD, and PPARG agonists on the expression of selected genes, apoptosis, and cell proliferation. The addition of CEM stimulated PPARA expression and DNA binding activity of this isoform in LE and ST cells (P < 0.05). Increased expression of PPARD mRNA in the presence of CEM was detected in ST cells (P < 0.05), while the concentration of PPARG transcripts decreased in response to CEM in both cell types (P < 0.05). LE and ST cells of the pig endometrium possess PPARA, PPARD, and PPARG proteins, with clear nuclear staining visible predominately in ST cells. In LE cells, activation of PPARG with 15-deoxy-Δ12,14-prostaglandin(PG)J2 down-regulated the expression of genes encoding amino acid transporter 1 (SLC38A1), leukemia inhibitory factor (LIF) and enzymes involved in PG synthesis (P < 0.05). In ST cells, activation of PPARD isoform with both agonists used (L-165,041 and cPGI2) and PPARG isoform with 15-deoxy-Δ12,14-PGJ2 increased vascular endothelial growth factor A (VEGFA) mRNA expression (P < 0.05). Moreover, GW9578 (PPARA agonist) and 15-deoxy-Δ12,14-PGJ2 stimulated glucose transporter 1 (SLC2A1) gene expression in ST cells. 15-deoxy-Δ12,14-PGJ2 was also effective in up-regulation of the ratio of BAX/BCL2 mRNA expression and active caspase-3 concentration in ST cells (P < 0.05). Finally, GW9578 stimulated LE and ST cell proliferation, while rosiglitazone (PPARG agonist) increased the number of viable ST but not LE cells. In conclusion, this study demonstrated that conceptus products differentially modulate PPARs expression and activity in the porcine endometrium. Activation of PPARs may in turn affect nutrient transport, PG synthesis, angiogenesis, apoptosis, or cell proliferation in this tissue. Therefore, PPAR isoforms seem to play an important role in development and function of the porcine uterus.


Assuntos
Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Compostos de Quinolínio/metabolismo , Suínos , Animais , Apoptose , Proliferação de Células , Endométrio/metabolismo , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , RNA Mensageiro/metabolismo , Células Estromais/metabolismo
11.
Sheng Li Xue Bao ; 70(5): 511-520, 2018 Oct 25.
Artigo em Chinês | MEDLINE | ID: mdl-30377690

RESUMO

Hypoxic exposure activates hypoxia inducible factors (HIFs) to up-regulate the expression of its target genes. These genes encode glucose metabolism related proteins, such as glucose transporters (GLUTs) and glycolysis related enzymes, including lactate dehydrogenase A (LDHA) and aldolase A (ALDA). Therefore, HIFs participate in oxygenolysis of glucose and play an important role in mediating hypoxia response and weight loss. Exercise training influences fatty acid metabolism, insulin sensitivity and body energy balance through activating peroxisome proliferator-activated receptors (PPARs), which plays an active role in losing weight. In addition, hypoxic exposure or exercise training can activate energy sensor 5'-AMP activated protein kinase (AMPK) in cells and promote oxidation of glucose and fatty acid and weight loss. It has been shown that hypoxic training exerts a better effects on controlling weight, compared with either hypoxic exposure or exercise training alone. This paper reviewed synergistic interactions among HIFs, PPARs and AMPK under hypoxic training and proposed possible mechanisms of hypoxic training-induced weight loss via AMPK-HIFs axis or AMPK-PPARs axis, thus providing theoretical guidance for application of hypoxic training in weight control.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Fator 1 Induzível por Hipóxia/fisiologia , Hipóxia , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Redução de Peso , Animais , Peso Corporal , Metabolismo Energético , Ácidos Graxos , Frutose-Bifosfato Aldolase/fisiologia , Glucose , Proteínas Facilitadoras de Transporte de Glucose/fisiologia , Humanos , Resistência à Insulina , Isoenzimas/fisiologia , L-Lactato Desidrogenase/fisiologia , Lactato Desidrogenase 5 , Metabolismo dos Lipídeos , Oxirredução , Regulação para Cima
12.
Gen Comp Endocrinol ; 268: 110-120, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30114400

RESUMO

Migratory birds undergo metabolic remodeling in tissues, including increased lipid storage in white adipose and fatty acid uptake and oxidation in skeletal muscle, to optimize energy substrate availability and utilization in preparation for long-distance flight. Different tissues undergo gene expression changes in keeping with their specialized functions and driven by tissue specific transcriptional pathways. Peroxisome proliferator-activated receptors (PPARs) are lipid-activated nuclear receptors that regulate metabolic pathways involved in lipid and glucose utilization or storage in mammals. To examine whether PPARs might mediate fatty acid activation of metabolic gene programs that would be relevant during pre-migratory fattening, we used gray catbird as the focal species. PPAR isoforms cloned from catbird share high amino acid identity with mammalian homologs (% vs human): gcPPARα (88.1%), gcPPARδ (87.3%), gcPPARγ (91.2%). We tested whether gcPPARs activated fatty acid (FA) utilization genes using Lpl and Cpt1b gene promoter-luciferase reporters in mammalian cell lines. In C2C12 mouse myocytes gcPPARα was broadly activated by the saturated and unsaturated FAs tested; while gcPPARδ showed highest activation by the mono-unsaturated FA, 18:1 oleic acid (+80%). In CV-1 monkey kidney cells gcPPARγ responded to the poly-unsaturated fatty acid, 20:5 eicosapentaenoic acid (+60%). Moreover, in agreement with their structural conservation, gcPPARs were activated by isoform selective synthetic agonists similar to the respective mammalian isoform. Adenoviral mediated over-expression of PPARα in C2C12 myocytes induced expression of genes involved in fatty acid transport, including Cd36/Fat, as well as Cpt1b, which mediates a key rate limiting step of mitochondrial ß-oxidation. These gene expression changes correlated with increased lipid droplet accumulation in C2C12 myoblasts and differentiated myotubes and enhanced ß-oxidation in myotubes. Collectively, the data predict that the PPARs play a conserved role in gray catbirds to regulate lipid metabolism in target tissues that undergo metabolic remodeling throughout the annual migratory cycle.


Assuntos
Ligantes , Metabolismo dos Lipídeos/fisiologia , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Ativação Transcricional/fisiologia , Animais , Aves , Diferenciação Celular/efeitos dos fármacos , Humanos
13.
Artigo em Inglês | MEDLINE | ID: mdl-30036119

RESUMO

Peroxisome proliferator-activated receptors (PPARs), as members of nuclear hormone receptor superfamily, can be activated by binding natural or synthetic ligands. The use of related ligands has revealed many potential roles for PPARs in the pathogenesis of some human metabolic disorders and inflammatory-related disease. Based on the previous studies, this review primarily concluded the current progress of knowledge regarding the specific biological activity of PPARs in cancers, atherosclerosis, and type 2 diabetes mellitus, providing a foundation for the potential therapeutic use of PPAR ligands in human diseases.


Assuntos
Aterosclerose/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Neoplasias/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Humanos
14.
Toxicol Sci ; 159(2): 436-448, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28962521

RESUMO

Skin tumorigenesis results from DNA damage, increased inflammation, and evasion of apoptosis. The peroxisome proliferator-activated receptors (PPARs) can modulate these mechanisms in non-melanoma skin cancer. However, limited data exists regarding the role of PPARs in melanoma. This study examined the effect of proliferator-activated receptor-ß/δ (PPARß/δ) and PPARγ on cell proliferation, anchorage-dependent clonogenicity, and ectopic xenografts in the UACC903 human melanoma cell line. Stable overexpression of either PPARß/δ or PPARγ enhanced ligand-induced expression of a PPARß/δ/PPARγ target gene in UACC903 cell lines as compared with controls. The induction of target gene expression by ligand activation of PPARγ was not altered by overexpression of PPARß/δ, or vice versa. Stable overexpression of either PPARß/δ or PPARγ reduced the percentage of cells in the G1 and S phase of the cell cycle, and increased the percentage of cells in the G2/M phase of the cell cycle in UACC903 cell lines as compared with controls. Ligand activation of PPARß/δ did not further alter the distribution of cells within each phase of the cell cycle. By contrast, ligand activation of PPARγ enhanced these changes in stable UACC903 cells overexpressing PPARγ compared with controls. Stable overexpression of either PPARß/δ or PPARγ and/or ligand activation of either PPARß/δ or PPARγ inhibited cell proliferation, and anchorage-dependent clonogenicity of UACC903 cell lines as compared with controls. Further, overexpression of either PPARß/δ or PPARγ and/or ligand activation of either PPARß/δ or PPARγ inhibited ectopic xenograft tumorigenicity derived from UACC903 melanoma cells as compared with controls, and this was likely due in part to induction of apoptosis. Results from these studies demonstrate the antitumorigenic effects of both PPARß/δ and PPARγ and suggest that targeting these receptors may be useful for primary or secondary melanoma chemoprevention.


Assuntos
Apoptose/fisiologia , Inflamação/fisiopatologia , Melanoma/patologia , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Neoplasias Cutâneas/patologia , Animais , Adesão Celular/fisiologia , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Xenoenxertos , Humanos , Ligantes , Camundongos , Camundongos Nus , Receptores Ativados por Proliferador de Peroxissomo/genética
15.
Am J Chin Med ; 45(6): 1147-1156, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28830207

RESUMO

The first record of ginseng use dates back over two millennia, and ginseng is now popular in more than 35 countries. Ginsenosides are the pharmacological constituents responsible for the beneficial effects of ginseng. There is increasing evidence that ginseng and its bioactive ingredients are involved in the regulation of nuclear receptors, molecules that act in response to the specific binding of hormones, which link to a diverse array of signaling pathways, such as the ERK and PI3K/Akt pathways. Knowledge of the mechanism of how ginseng mediates these complexes is essential for the development of multi-target phytomedicine as possible therapy for different diseases. Here, we discuss the literature on the effects of ginseng and its constituents on estrogen, glucocorticoid, peroxisome proliferator-activated, and androgen nuclear hormone receptors, as well as how ginseng and its constituents exert their biological function in the treatment of cancer, obesity, and cardiovascular and neurological disorders. The accumulated results definitely show that the nuclear receptors are cellular targets of ginsenosides, but more rigorous data are required to establish and provide a scientific basis to confirm the suggested efficacy of ginseng or products with ginsenosides.


Assuntos
Ginsenosídeos/farmacologia , Ginsenosídeos/uso terapêutico , Panax/química , Fitoterapia , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Animais , Doenças Cardiovasculares/tratamento farmacológico , Feminino , Ginsenosídeos/isolamento & purificação , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Neoplasias/tratamento farmacológico , Doenças do Sistema Nervoso/tratamento farmacológico , Obesidade/tratamento farmacológico , Receptores Ativados por Proliferador de Peroxissomo/efeitos dos fármacos , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Extratos Vegetais/isolamento & purificação , Receptores Androgênicos/efeitos dos fármacos , Receptores Androgênicos/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/fisiologia , Receptores de Glucocorticoides/efeitos dos fármacos , Receptores de Glucocorticoides/fisiologia
17.
Biochimie ; 136: 3-11, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28034718

RESUMO

Oxylipins are bioactive metabolites derived from the oxygenation of ω3 and ω6 polyunsaturated fatty acids, triggered essentially by cyclooxygenase and lipoxygenase activities. Oxylipins are involved in the development and function of adipose tissue and their productions are strictly related to diet quality and quantity. Oxylipins signal via cell surface membrane (G Protein-coupled receptors) and nuclear receptors (peroxisome proliferator-activated receptors), two pathways playing a pivotal role in adipocyte biology. In this review, we made an attempt to cover the available knowledge about synthesis and molecular function of oxylipins known to modulate adipogenesis, adipocyte function and phenotype conversion, with a focus on their interaction with peroxisome proliferator-activated nuclear receptor family.


Assuntos
Adipogenia/fisiologia , Oxilipinas/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Humanos
18.
PLoS One ; 11(9): e0162228, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27611793

RESUMO

Adipocytes are differentiated by various transcriptional cascades integrated on the master regulator, Pparγ. To discover new genes involved in adipocyte differentiation, preadipocytes were treated with three newly identified pro-adipogenic small molecules and GW7845 (a Pparγ agonist) for 24 hours and transcriptional profiling was analyzed. Four genes, Peroxisome proliferator-activated receptor γ (Pparγ), human complement factor D homolog (Cfd), Chemokine (C-C motif) ligand 9 (Ccl9), and GIPC PDZ Domain Containing Family Member 2 (Gipc2) were induced by at least two different small molecules but not by GW7845. Cfd and Ccl9 expressions were specific to adipocytes and they were altered in obese mice. Small hairpin RNA (shRNA) mediated knockdown of Cfd in preadipocytes inhibited lipid accumulation and expression of adipocyte markers during adipocyte differentiation. Overexpression of Cfd promoted adipocyte differentiation, increased C3a production, and led to induction of C3a receptor (C3aR) target gene expression. Similarly, treatments with C3a or C3aR agonist (C4494) also promoted adipogenesis. C3aR knockdown suppressed adipogenesis and impaired the pro-adipogenic effects of Cfd, further suggesting the necessity for C3aR signaling in Cfd-mediated pro-adipogenic axis. Together, these data show the action of Cfd in adipogenesis and underscore the application of small molecules to identify genes in adipocytes.


Assuntos
Adipogenia/genética , Fator D do Complemento/fisiologia , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Adipogenia/fisiologia , Animais , Linhagem Celular , Complemento C3a/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxazóis/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Receptores de Complemento/metabolismo , Transdução de Sinais , Bibliotecas de Moléculas Pequenas , Transcriptoma , Tirosina/análogos & derivados , Tirosina/farmacologia
19.
Int J Obes (Lond) ; 40(11): 1768-1775, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27599633

RESUMO

OBJECTIVE: Although intrauterine nutritional stress is known to result in offspring obesity and the metabolic phenotype, the underlying cellular/molecular mechanisms remain incompletely understood. We tested the hypothesis that compared with the controls, the bone marrow-derived mesenchymal stem cells (BMSCs) of the intrauterine growth-restricted (IUGR) offspring exhibit a more adipogenic phenotype. METHODS: A well-established rat model of maternal food restriction (MFR), that is, 50% global caloric restriction during the later-half of pregnancy and ad libitum diet following birth that is known to result in an obese offspring with a metabolic phenotype was used. BMSCs at 3 weeks of age were isolated, and then molecularly and functionally profiled. RESULTS: BMSCs of the intrauterine nutritionally-restricted offspring demonstrated an increased proliferation and an enhanced adipogenic molecular profile at miRNA, mRNA and protein levels, with an overall up-regulated PPARγ (miR-30d, miR-103, PPARγ, C/EPBα, ADRP, LPL, SREBP1), but down-regulated Wnt (LRP5, LEF-1, ß-catenin, ZNF521 and RUNX2) signaling profile. Following adipogenic induction, compared with the control BMSCs, the already up-regulated adipogenic profile of the MFR BMSCs, showed a further increased adipogenic response. CONCLUSIONS: Markedly enhanced adipogenic molecular profile and increased cell proliferation of MFR BMSCs suggest a possible novel cellular/mechanistic link between the intrauterine nutritional stress and offspring metabolic phenotype. This provides new potential predictive and therapeutic targets against these conditions in the IUGR offspring.


Assuntos
Adipogenia/fisiologia , Retardo do Crescimento Fetal/patologia , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Células-Tronco Mesenquimais/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Animais Recém-Nascidos , Restrição Calórica , Diferenciação Celular , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Retardo do Crescimento Fetal/genética , Fenômenos Fisiológicos da Nutrição Materna/genética , MicroRNAs , Fenótipo , Gravidez , RNA Mensageiro , Ratos , Ratos Sprague-Dawley , Regulação para Cima
20.
J Diabetes Investig ; 7(2): 179-89, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27042269

RESUMO

AIMS/INTRODUCTION: Uncoupling protein 2 (UCP2), which was an important mitochondrial inner membrane protein associated with glucose and lipid metabolism, widely expresses in all kinds of tissues including hepatocytes. The present study aimed to explore the impact of UCP2 deficiency on glucose and lipid metabolism, insulin sensitivity and its effect on the liver-associated signaling pathway by expression profiling analysis. MATERIALS AND METHODS: Four-week-old male UCP2-/- mice and UCP2+/+ mice were randomly assigned to four groups: UCP2-/- on a high-fat diet, UCP2-/- on a normal chow diet, UCP2+/+ on a high-fat diet and UCP2+/+ on a normal chow diet. The differentially expressed genes in the four groups on the 16th week were identified by Affymetrix gene array. RESULTS: The results of intraperitoneal glucose tolerance test and insulin tolerance showed that blood glucose and ß-cell function were improved in the UCP2-/- group on high-fat diet. Enhanced insulin sensitivity was observed in the UCP2-/- group. The differentially expressed genes were mapped to 23 pathways (P < 0.05). We concentrated on the 'peroxisome proliferator-activated receptor (PPAR) signaling pathway' (P = 3.19 × 10(-11)), because it is closely associated with the regulation of glucose and lipid profiles. In the PPAR signaling pathway, seven genes (PPARγ, Dbi, Acsl3, Lpl, Me1, Scd1, Fads2) in the UCP2-/- mice were significantly upregulated. CONCLUSIONS: The present study used gene arrays to show that activity of the PPAR signaling pathway involved in the improvement of glucose and lipid metabolism in the liver of UCP2-deficient mice on a long-term high-fat diet. The upregulation of genes in the PPAR signaling pathway could explain our finding that UCP2 deficiency ameliorated insulin sensitivity. The manipulation of UCP2 protein expression could represent a new strategy for the prevention and treatment of diabetes.


Assuntos
Glucose/metabolismo , Proteína Desacopladora 2/fisiologia , Animais , Glicemia , Dieta Hiperlipídica , Perfilação da Expressão Gênica , Teste de Tolerância a Glucose , Resistência à Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Metabolismo dos Lipídeos , Fígado/metabolismo , Masculino , Camundongos , Receptores Ativados por Proliferador de Peroxissomo/genética , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Distribuição Aleatória , Transdução de Sinais , Proteína Desacopladora 2/genética , Proteína Desacopladora 2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA