Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Environ Sci Technol ; 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39382118

RESUMEN

Cresyl diphenyl phosphate (CDP), a novel organophosphate ester (OPE), has been detected in various environmental and human samples. However, there is very limited knowledge regarding its toxicity, mechanisms of action, and potential health risks. Using new alternative methods (NAMs), across the molecular interactions, signaling pathways, cell functions, animal effects, and population risks, we investigated the potential adipogenic effects and associated risks of CDP and legacy OPE triphenyl phosphate (TPHP) by acting on peroxisome proliferator-activated receptor gamma (PPARγ). Among the 19 screened OPEs, CDP bound to PPARγ with the highest binding potency, followed by TPHP. CDP activated PPARγ through fitting into the binding pocket with strong hydrophobicity and hydrogen bond interactions; CDP exhibited higher potency compared to TPHP. In 3T3-L1 cells, CDP enhanced the PPARγ-mediated adipogenesis activity, exhibiting greater potency than TPHP. The intracellular concentration and receptor-bound concentrations (RBC) of CDP were also higher than those of TPHP in both HEK293 cells and 3T3-L1 cells. In mice, exposure to CDP activated the PPARγ-mediated adipogenic pathway, leading to an increased white adipose tissue weight gain. Overall, CDP could bind to and activate PPARγ, thereby promoting preadipocyte differentiation and the development of white adipose tissue. Its potential obesogenic risks should be of high concern.

2.
Adv Exp Med Biol ; 1460: 97-129, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39287850

RESUMEN

The ratio of free fatty acid (FFA) turnover decreases significantly with the expansion of white adipose tissue. Adipose tissue and dietary saturated fatty acid levels significantly correlate with an increase in fat cell size and number. The G0/G1 switch gene 2 increases lipid content in adipocytes and promotes adipocyte hypertrophy through the restriction of triglyceride (triacylglycerol: TAG) turnover. Hypoxia in obese adipose tissue due to hypertrophic adipocytes results in excess deposition of extracellular matrix (ECM) components. Cluster of differentiation (CD) 44, as the main receptor of the extracellular matrix component regulates cell-cell and cell-matrix interactions including diet-induced insulin resistance. Excess TAGs, sterols, and sterol esters are surrounded by the phospholipid monolayer surface and form lipid droplets (LDs). Once LDs are formed, they grow up because of the excessive amount of intracellular FFA stored and reach a final size. The ratio of FFA turnover/lipolysis decreases significantly with increases in the degree of obesity. Dysfunctional adipose tissue is unable to expand further to store excess dietary lipids, increased fluxes of plasma FFAs lead to ectopic fatty acid deposition and lipotoxicity. Reduced neo-adipogenesis and dysfunctional lipid-overloaded adipocytes are hallmarks of hypertrophic obesity linked to insulin resistance. Obesity-associated adipocyte death exhibits feature of necrosis-like programmed cell death. Adipocyte death is a prerequisite for the transition from hypertrophic to hyperplastic obesity. Increased adipocyte number in obesity has life-long effects on white adipose tissue mass. The positive correlation between the adipose tissue volume and magnetic resonance imaging proton density fat fraction estimation is used for characterization of the obesity phenotype, as well as the risk stratification and selection of appropriate treatment strategies. In obese patients with type 2 diabetes, visceral adipocytes exposed to chronic/intermittent hyperglycemia develop a new microRNAs' (miRNAs') expression pattern. Visceral preadipocytes memorize the effect of hyperglycemia via changes in miRNAs' expression profile and contribute to the progression of diabetic phenotype. Nonsteroidal anti-inflammatory drugs, metformin, and statins can be beneficial in treating the local or systemic consequences of white adipose tissue inflammation. Rapamycin inhibits leptin-induced LD formation. Collectively, in this chapter, the concept of adipose tissue remodeling in response to adipocyte death or adipogenesis, and the complexity of LD interactions with the other cellular organelles are reviewed. Furthermore, clinical perspective of fat cell turnover in obesity is also debated.


Asunto(s)
Adipocitos , Lipólisis , Obesidad , Humanos , Obesidad/metabolismo , Obesidad/patología , Adipocitos/metabolismo , Adipocitos/patología , Animales , Metabolismo de los Lípidos , Adipogénesis , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Resistencia a la Insulina
3.
Adv Exp Med Biol ; 1390: 123-141, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36107316

RESUMEN

Nuclear receptors (NRs) are transcription factors that modulate gene expression in a ligand-dependent manner. The ubiquitously expressed glucocorticoid receptor (GR) and peroxisome proliferator-activated receptor gamma (PPARγ) represent steroid (type I) and non-steroid (type II) classes of NRs, respectively. The diverse transcriptional and physiological outcomes of their activation are highly tissue-specific. For example, in subsets of immune cells, such as macrophages, the signaling of GR and PPARγ converges to elicit an anti-inflammatory phenotype; in contrast, in the adipose tissue, their signaling can lead to reciprocal metabolic outcomes. This review explores the cooperative and divergent outcomes of GR and PPARγ functions in different cell types and tissues, including immune cells, adipose tissue and the liver. Understanding the coordinated control of these NR pathways should advance studies in the field and potentially pave the way for developing new therapeutic approaches to exploit the GR:PPARγ crosstalk.


Asunto(s)
PPAR gamma , Receptores de Glucocorticoides , Antiinflamatorios/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Ligandos , PPAR gamma/genética , Receptores de Glucocorticoides/genética , Factores de Transcripción/fisiología
4.
J Biol Chem ; 295(33): 11866-11876, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32616652

RESUMEN

Garcinoic acid (GA or δ-T3-13'COOH), is a natural vitamin E metabolite that has preliminarily been identified as a modulator of nuclear receptors involved in ß-amyloid (Aß) metabolism and progression of Alzheimer's disease (AD). In this study, we investigated GA's effects on Aß oligomer formation and deposition. Specifically, we compared them with those of other vitamin E analogs and the soy isoflavone genistein, a natural agonist of peroxisome proliferator-activated receptor γ (PPARγ) that has therapeutic potential for managing AD. GA significantly reduced Aß aggregation and accumulation in mouse cortical astrocytes. Similarly to genistein, GA up-regulated PPARγ expression and apolipoprotein E (ApoE) efflux in these cells with an efficacy that was comparable with that of its metabolic precursor δ-tocotrienol and higher than those of α-tocopherol metabolites. Unlike for genistein and the other vitamin E compounds, the GA-induced restoration of ApoE efflux was not affected by pharmacological inhibition of PPARγ activity, and specific activation of pregnane X receptor (PXR) was observed together with ApoE and multidrug resistance protein 1 (MDR1) membrane transporter up-regulation in both the mouse astrocytes and brain tissue. These effects of GA were associated with reduced Aß deposition in the brain of TgCRND8 mice, a transgenic AD model. In conclusion, GA holds potential for preventing Aß oligomerization and deposition in the brain. The mechanistic aspects of GA's properties appear to be distinct from those of other vitamin E metabolites and of genistein.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Benzopiranos/farmacología , Encéfalo/efectos de los fármacos , Agregación Patológica de Proteínas/prevención & control , Vitamina E/análogos & derivados , Péptidos beta-Amiloides/ultraestructura , Animales , Benzopiranos/farmacocinética , Encéfalo/metabolismo , Encéfalo/patología , Masculino , Ratones , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/patología , Vitamina E/farmacocinética , Vitamina E/farmacología
5.
Lipids Health Dis ; 20(1): 39, 2021 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-33879188

RESUMEN

BACKGROUND: To investigate the roles of the transcription factors twist family bHLH transcription factor 1 (TWIST1), twist family bHLH transcription factor 2 (TWIST2), and peroxisome proliferator activated receptor gamma (PPARγ) in the progression of nonalcoholic steatohepatitis. METHODS: The protein levels of TWIST1, TWIST2 and PPARγ were determined in the serum of nonalcoholic fatty liver disease (NAFLD) patients and healthy controls by enzyme-linked immunosorbent assay (ELISA). An in vivo model for fatty liver was established by feeding C57BL/6 J mice a high-fat diet (HFD). An in vitro model of steatosis was established by treating LO-2 cells with oleic acid (OA). RNA sequencing was performed on untreated and OA-treated LO-2 cells followed by TWIST1, TWIST2 and PPARγ gene mRNA levels analysis, Gene Ontology (GO) enrichment and pathway analysis. RESULTS: The TWIST2 serum protein levels decreased significantly in all fatty liver groups (P < 0.05), while TWIST1 varied. TWIST2 tended to be lower in mice fed an HFD and was significantly lower at 3 months. Similarly, in the in vitro model, the TWIST2 protein level was downregulated significantly at 48 and 72 h after OA treatment. RNA sequencing of LO-2 cells showed an approximately 2.3-fold decrease in TWIST2, with no obvious change in TWIST1 and PPARγ. The PPAR signaling pathway was enriched, with 4 genes upregulated in OA-treated cells (P = 0.0018). The interleukin (IL)-17 and tumor necrosis factor (TNF) signaling pathways were enriched in OA-treated cells. CONCLUSIONS: The results provide evidence that the TWIST2 and PPAR signaling pathways are important in NAFLD and shed light on a potential mechanism of steatosis.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico/metabolismo , PPAR gamma/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Proteína 1 Relacionada con Twist/metabolismo , Adolescente , Adulto , Animales , Western Blotting , Estudios de Casos y Controles , Línea Celular , Notificación de Enfermedades , Progresión de la Enfermedad , Femenino , Prueba de Tolerancia a la Glucosa , Hepatocitos/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/patología , Proteínas Nucleares/sangre , Proteínas Nucleares/metabolismo , PPAR gamma/sangre , Proteínas Represoras/sangre , Proteína 1 Relacionada con Twist/sangre , Adulto Joven
6.
Int J Mol Sci ; 22(16)2021 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-34445309

RESUMEN

In our previous work, we built the model of PPARγ dependent pathways involved in the development of the psoriatic lesions. Peroxisome proliferator-activated receptor gamma (PPARγ) is a nuclear receptor and transcription factor which regulates the expression of many proinflammatory genes. We tested the hypothesis that low levels of PPARγ expression promote the development of psoriatic lesions triggering the IL17-related signaling cascade. Skin samples of normally looking and lesional skin donated by psoriasis patients and psoriatic CD3+ Tcells samples (n = 23) and samples of healthy CD3+ T cells donated by volunteers (n = 10) were analyzed by real-time PCR, ELISA and immunohistochemistry analysis. We found that the expression of PPARγ is downregulated in human psoriatic skin and laser treatment restores the expression. The expression of IL17, STAT3, FOXP3, and RORC in psoriatic skin before and after laser treatment were correlated with PPARγ expression according to the reconstructed model of PPARγ pathway in psoriasis.In conclusion, we report that PPARγ weakens the expression of genes that contribute in the development of psoriatic lesion. Our data show that transcriptional regulation of PPARγ expression by FOSL1 and by STAT3/FOSL1 feedback loop may be central in the psoriatic skin and T-cells.


Asunto(s)
PPAR gamma/metabolismo , Psoriasis/metabolismo , Transducción de Señal , Adulto , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Interleucina-17/metabolismo , Masculino , Persona de Mediana Edad , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , PPAR gamma/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factor de Transcripción STAT3/metabolismo , Linfocitos T/metabolismo
7.
Molecules ; 25(5)2020 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-32138197

RESUMEN

Phytocannabinoids (pCBs) are a large family of meroterpenoids isolated from the plant Cannabis sativa. Δ9-Tetrahydrocannabinol (THC) and cannabidiol (CBD) are the best investigated phytocannabinoids due to their relative abundance and interesting bioactivity profiles. In addition to various targets, THC and CBD are also well-known agonists of peroxisome proliferator-activated receptor gamma (PPARγ), a nuclear receptor involved in energy homeostasis and lipid metabolism. In the search of new pCBs potentially acting as PPARγ agonists, we identified cannabimovone (CBM), a structurally unique abeo-menthane pCB, as a novel PPARγ modulator via a combined computational and experimental approach. The ability of CBM to act as dual PPARγ/α agonist was also evaluated. Computational studies suggested a different binding mode toward the two isoforms, with the compound able to recapitulate the pattern of H-bonds of a canonical agonist only in the case of PPARγ. Luciferase assays confirmed the computational results, showing a selective activation of PPARγ by CBM in the low micromolar range. CBM promoted the expression of PPARγ target genes regulating the adipocyte differentiation and prevented palmitate-induced insulin signaling impairment. Altogether, these results candidate CBM as a novel bioactive compound potentially useful for the treatment of insulin resistance-related disorders.


Asunto(s)
Cannabinoides/química , Cannabinoides/farmacología , Cannabis/química , PPAR gamma/agonistas , PPAR gamma/metabolismo , Células 3T3-L1 , Animales , Metabolismo Energético/efectos de los fármacos , Concentración de Iones de Hidrógeno , Resistencia a la Insulina/fisiología , Ratones , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
8.
J Cell Biochem ; 119(9): 7729-7737, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29923224

RESUMEN

The agonists of peroxisome proliferator-activated receptor gamma (PPARγ) from natural victual products were used as antidiabetic agents. Faba bean (Vicia faba L.) is a consequential legume that was known to possess potential antidiabetic activity, whose mechanism of action was unknown. The current study was focused to ascertain gene expression of the nuclear receptor PPARγ by Faba bean pod extract in rat cell lines (RINm5F).The real-time polymerase chain reaction analysis demonstrated that Faba bean pod extract in concentrations of 160 µg/mL have shown 4.97-fold stimulation compared with control. The cells treated with 320 µg/mL has shown 5.89-fold upregulation, respectively. Furthermore, in silico docking analysis was carried out against PPARγ, using the bioactive compounds identified from Faba bean pod extracts, which were known reported compounds from the literature. The results suggest that gene expression of PPARγ was inhibited by the constituents in Faba bean. In silico analysis prognosticates, butein has a high binding energy (-8.6 kcal/mol) with an atomic contact energy of -214.10, followed by Apigenin and Quercetin against PPARγ. Similarly, the percentage of interaction was high for butein, followed by Apigenin and Quercetin than other compounds comparatively. Hence, the results conclude inhibition of PPARγ by the bioactive compounds from Faba bean, which may provide insights into developing future therapeutic molecules for diabetes mellitus.


Asunto(s)
Línea Celular/química , Hipoglucemiantes/farmacología , PPAR gamma/genética , Extractos Vegetales/farmacología , Regulación hacia Arriba , Animales , Apigenina/química , Apigenina/farmacología , Chalconas/química , Chalconas/farmacología , Simulación por Computador , Relación Dosis-Respuesta a Droga , Hipoglucemiantes/química , Modelos Biológicos , Modelos Moleculares , Simulación del Acoplamiento Molecular , PPAR gamma/agonistas , PPAR gamma/química , Extractos Vegetales/química , Quercetina/química , Ratas , Vicia faba
9.
Biochem Biophys Res Commun ; 504(1): 68-74, 2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-30173890

RESUMEN

Reverse cholesterol transport (RCT) plays an important role in cholesterol and lipid metabolism. Regulating the activities of key transporters and receptors in RCT, such as ATP-binding cassette transporter A1 (ABCA1), helps to prevent atherosclerotic cardiovascular disease. In this study, we used an ABCA1 promoter luciferase reporter assay to screen 20,000 compounds for ABCA1 upregulators. Compound E3317 (N-(6-butylbenzo[d]thiazol-2(3H)-ylidene)-3-(N-(2-cyanoethyl)sulfamoyl)benzamide)) was identified as a positive hit with an EC50 value of 0.2 µM in ABCA1p-LUC HepG2 cells. Thus, we hypothesized that E3317 might have cholesterol- and lipid metabolism-regulating effects through ABCA1 upregulation. E3317 significantly increased ABCA1 mRNA and protein expression in hepatic L02 cells and RAW264.7 macrophages. E3317 promoted cholesterol efflux to apolipoprotein A-I in RAW264.7 macrophages and significantly decreased lipid accumulation in oxidized low-density lipoprotein-induced murine RAW264.7 macrophages. Further studies using ABCA1 siRNA showed that the promotion of cholesterol efflux and decrease of lipid accumulation by E3317 depended on ABCA1 expression. Mechanistic studies indicated that E3317 regulated ABCA1 expression via activating nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ), which plays an important role in the regulation of glucose homeostasis and lipid metabolism. The structure of E3317 was docked in the ligand-binding domain of PPARγ (PBD code: 4EMA) to find the key binding amino acids. Site mutation assays confirmed that Y327 and F363 were the key PPARγ binding epitopes of E3317. Our results revealed that E3317 upregulates ABCA1 expression and thereby promotes cholesterol efflux. E3317 may regulate ABCA1 expression through PPARγ. Our findings provide a new compound, E3317, which may have beneficial cardiovascular effects.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/biosíntesis , Colesterol/metabolismo , Antagonistas Colinérgicos/farmacología , Transportador 1 de Casete de Unión a ATP/genética , Animales , Transporte Biológico/efectos de los fármacos , Línea Celular , Antagonistas Colinérgicos/química , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , PPAR gamma/química , Dominios Proteicos , Células RAW 264.7 , Activación Transcripcional/efectos de los fármacos
10.
Mov Disord ; 32(11): 1594-1599, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28861893

RESUMEN

BACKGROUND: Whether antidiabetic glitazone drugs protect against Parkinson's disease remains controversial. Although a single clinical trial showed no evidence of disease modulation, retrospective studies suggest that a disease-preventing effect may be plausible. The objective of this study was to examine if the use of glitazone drugs is associated with a lower incidence of PD among diabetic patients. METHODS: We compared the incidence of PD between individuals with diabetes who used glitazones, with or without metformin, and individuals using only metformin in the Norwegian Prescription Database. This database contains all prescription drugs dispensed for the entire Norwegian population. We identified 94,349 metformin users and 8396 glitazone users during a 10-year period and compared the incidence of PD in the 2 groups using Cox regression survival analysis, with glitazone exposure as a time-dependent covariate. RESULTS: Glitazone use was associated with a significantly lower incidence of PD compared with metformin-only use (hazard ratio, 0.72; 95% confidence interval, 0.55-0.94; P = 0.01). CONCLUSIONS: The use of glitazones is associated with a decreased risk of incident PD in populations with diabetes. Further studies are warranted to confirm and understand the role of glitazones in neurodegeneration. © 2017 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Metformina/farmacología , Enfermedad de Parkinson/prevención & control , Tiazolidinedionas/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Bases de Datos Factuales/estadística & datos numéricos , Diabetes Mellitus Tipo 2/epidemiología , Prescripciones de Medicamentos/estadística & datos numéricos , Quimioterapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Noruega/epidemiología , Enfermedad de Parkinson/epidemiología
11.
J Neuroinflammation ; 13(1): 145, 2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27287266

RESUMEN

BACKGROUND: A characteristic phenotype of congenital generalized lipodystrophy 2 (CGL2) that is caused by loss-of-function of seipin gene is mental retardation. Seipin is highly expressed in hippocampal pyramidal cells and astrocytes. Neuronal knockout of seipin in mice (seipin-KO mice) reduces the hippocampal peroxisome proliferator-activated receptor gamma (PPARγ) level without the loss of pyramidal cells. The down-regulation of PPARγ has gained increasing attention in neuroinflammation of Alzheimer's disease (AD). Thus, the present study focused on exploring the influence of seipin depletion on ß-amyloid (Aß)-induced neuroinflammation and Aß neurotoxicity. METHODS: Adult male seipin-KO mice were treated with a single intracerebroventricular (i.c.v.) injection of Aß25-35 (1.2 nmol/mouse) or Aß1-42 (0.1 nmol/mouse), generally a non-neurotoxic dose in wild-type (WT) mice. Spatial cognitive behaviors were assessed by Morris water maze and Y-maze tests, and hippocampal CA1 pyramidal cells and inflammatory responses were examined. RESULTS: The Aß25-35/1-42 injection in the seipin-KO mice caused approximately 30-35 % death of pyramidal cells and production of Hoechst-positive cells with the impairment of spatial memory. In comparison with the WT mice, the number of astrocytes and microglia in the seipin-KO mice had no significant difference, whereas the levels of IL-6 and TNF-α were slightly increased. Similarly, the Aß25-35/1-42 injection in the seipin-KO mice rather than the WT mice could stimulate the activation of astrocytes or microglia and further elevated the levels of IL-6 and TNF-α. Treatment of the seipin-KO mice with the PPARγ agonist rosiglitazone (rosi) could prevent Aß25-35/1-42-induced neuroinflammation and neurotoxicity, which was blocked by the PPARγ antagonist GW9962. In the seipin-KO mice, the level of glycogen synthase kinase-3ß (GSK3ß) phosphorylation at Tyr216 was elevated, while at Ser9, it was reduced compared to the WT mice, which were corrected by the rosi treatment but were unaffected by the Aß25-35 injection. CONCLUSIONS: Seipin deficiency in astrocytes increases GSK3ß activity and levels of IL-6 and TNF-α through reducing PPARγ, which can facilitate Aß25-35/1-42-induced neuroinflammation to cause the death of neuronal cells and cognitive deficits.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Encefalitis , Proteínas de Unión al GTP Heterotriméricas/deficiencia , Hipocampo/metabolismo , Síndromes de Neurotoxicidad , PPAR gamma/metabolismo , Fragmentos de Péptidos/toxicidad , Animales , Proteínas de Unión al Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Encefalitis/etiología , Encefalitis/genética , Encefalitis/patología , Subunidades gamma de la Proteína de Unión al GTP , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteínas de Unión al GTP Heterotriméricas/genética , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/patología
12.
Pharmacol Res ; 111: 76-85, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27268145

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated nuclear receptor that regulates glucose and lipid metabolism, endothelial function and inflammation. Rosiglitazone (RGZ) and other thiazolidinedione (TZD) synthetic ligands of PPARγ are insulin sensitizers that have been used for the treatment of type 2 diabetes. However, undesirable side effects including weight gain, fluid retention, bone loss, congestive heart failure, and a possible increased risk of myocardial infarction and bladder cancer, have limited the use of TZDs. Therefore, there is a need to better understand PPARγ signaling and to develop safer and more effective PPARγ-directed therapeutics. In addition to PPARγ itself, many PPARγ ligands including TZDs bind to and activate G protein-coupled receptor 40 (GPR40), also known as free fatty acid receptor 1. GPR40 signaling activates stress kinase pathways that ultimately regulate downstream PPARγ responses. Recent studies in human endothelial cells have demonstrated that RGZ activation of GPR40 is essential to the optimal propagation of PPARγ genomic signaling. RGZ/GPR40/p38 MAPK signaling induces and activates PPARγ co-activator-1α, and recruits E1A binding protein p300 to the promoters of target genes, markedly enhancing PPARγ-dependent transcription. Therefore in endothelium, GPR40 and PPARγ function as an integrated signaling pathway. However, GPR40 can also activate ERK1/2, a proinflammatory kinase that directly phosphorylates and inactivates PPARγ. Thus the role of GPR40 in PPARγ signaling may have important implications for drug development. Ligands that strongly activate PPARγ, but do not bind to or activate GPR40 may be safer than currently approved PPARγ agonists. Alternatively, biased GPR40 agonists might be sought that activate both p38 MAPK and PPARγ, but not ERK1/2, avoiding its harmful effects on PPARγ signaling, insulin resistance and inflammation. Such next generation drugs might be useful in treating not only type 2 diabetes, but also diverse chronic and acute forms of vascular inflammation such as atherosclerosis and septic shock.


Asunto(s)
Fármacos Cardiovasculares/uso terapéutico , Hipoglucemiantes/uso terapéutico , PPAR gamma/agonistas , Transducción de Señal/efectos de los fármacos , Animales , Fármacos Cardiovasculares/efectos adversos , Diseño de Fármacos , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ácidos Grasos/metabolismo , Humanos , Hipoglucemiantes/efectos adversos , Ligandos , Terapia Molecular Dirigida , Óxido Nítrico/metabolismo , PPAR gamma/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Gen Comp Endocrinol ; 238: 61-68, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27131391

RESUMEN

Obesity has reached pandemic proportions, and there is mounting evidence that environmental exposures to endocrine disrupting chemicals known as "obesogens" may contribute to obesity and associated medical conditions. The Deepwater Horizon (DWH) oil spill resulted in a massive environmental release of crude oil and remediation efforts applied large quantities of Corexit dispersants to the oil spill. The Corexit-enhanced Water Accommodated Fraction (CWAF) of DWH crude oil contains PPARγ transactivation activity, which is attributed to dioctyl sodium sulfosuccinate (DOSS), a probable obesogen. In addition to its use in oil dispersants, DOSS is commonly used as a stool softener and food additive. Because PPARγ functions as a heterodimer with RXRα to transcriptionally regulate adipogenesis we investigated the potential of CWAF to transactivate RXRα and herein demonstrated that the Corexit component Span 80 has RXRα transactivation activity. Span 80 bound to RXRα in the low micromolar range and promoted adipocyte differentiation of 3T3-L1 preadipocytes. Further, the combination of DOSS and Span 80 increased 3T3-L1 adipocyte differentiation substantially more than treatment with either chemical individually, likely increasing the obesogenic potential of Corexit dispersants. From a public health standpoint, the use of DOSS and Span 80 as food additives heightens concerns regarding their use and mandates further investigations.


Asunto(s)
Emulsionantes/farmacología , Alimentos , Hexosas/farmacología , Obesidad/patología , Contaminación por Petróleo , Tensoactivos/farmacología , Activación Transcripcional/genética , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Animales , Ácido Dioctil Sulfosuccínico/farmacología , Células HEK293 , Humanos , Ratones , Ácido Oléico/farmacología , PPAR gamma/genética , Petróleo , Receptor alfa X Retinoide/genética , Activación Transcripcional/efectos de los fármacos
14.
Phytother Res ; 29(2): 281-7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25366162

RESUMEN

Saponins are a diverse group of biologically functional products in plants. Soyasaponins are usually glycosylated, which give rise to a wide diversity of structures and functions. In this study, we investigated the effects and molecular mechanism of soyasaponins Aa and Ab in regulating adipocyte differentiation and expression of adipogenic marker genes in 3T3-L1 adipocytes. Soyasaponins Aa and Ab dose-dependently inhibited the accumulation of lipids and the expression of adiponectin, adipocyte determination and differentiation factor 1/sterol regulatory element binding protein 1c, adipocyte fatty acid-binding protein 2, fatty acid synthase, and resistin in 3T3-L1 adipocytes. In addition, soyasaponins Aa and Ab suppressed the transcriptional activity of peroxisome proliferator-activated receptor γ (PPARγ) in HEK 293T cells. Furthermore, we confirmed that the expression of PPARγ and of CCAAT-enhancer-binding protein α (C/EBPα) was suppressed at both the mRNA and protein levels in 3T3-L1 adipocytes by treatment with soyasaponins Aa and Ab. Taken together, these findings indicate that soyasaponin Aa and Ab markedly inhibit adipocyte differentiation and expression of various adipogenic marker genes through the downregulation of the adipogenesis-related transcription factors PPARγ and C/EBPα in 3T3-L1 adipocytes.


Asunto(s)
Adipocitos/efectos de los fármacos , Fármacos Antiobesidad/farmacología , PPAR gamma/metabolismo , Saponinas/farmacología , Células 3T3-L1 , Adipogénesis/efectos de los fármacos , Animales , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Ratones
15.
Am J Physiol Lung Cell Mol Physiol ; 306(4): L361-71, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24337925

RESUMEN

Increased endothelin-1 (ET-1) disrupts angiogenesis in persistent pulmonary hypertension of the newborn (PPHN), but pathogenic mechanisms are unclear. Peroxisome proliferator activated receptor γ (PPARγ) is decreased in adult pulmonary hypertension, but whether ET-1-PPARγ interactions impair endothelial cell function and angiogenesis in PPHN remains unknown. We hypothesized that increased PPHN pulmonary artery endothelial cell (PAEC) ET-1 production decreases PPARγ signaling and impairs tube formation in vitro. Proximal PAECs were harvested from fetal sheep after partial ligation of the ductus arteriosus in utero (PPHN) and controls. PPARγ and phospho-PPARγ protein were compared between normal and PPHN PAECs ± ET-1 and bosentan (ETA/ETB receptor blocker). Tube formation was assessed in response to PPARγ agonists ± ET-1, N-nitro-l-arginine (LNA) (NOS inhibitor), and PPARγ siRNA. Endothelial NO synthase (eNOS), phospho-eNOS, and NO production were measured after exposure to PPARγ agonists and PPARγ siRNA. At baseline, PPHN PAECs demonstrate decreased tube formation and PPARγ protein expression and activity. PPARγ agonists restored PPHN tube formation to normal. ET-1 decreased normal and PPHN PAEC tube formation, which was rescued by PPARγ agonists. ET-1 decreased PPARγ protein and activity, which was prevented by bosentan. PPARγ agonists increased eNOS protein and activity and NO production in normal and PPHN PAECs. LNA inhibited the effect of PPARγ agonists on tube formation. PPARγ siRNA decreased eNOS protein and tube formation in normal PAECs. We conclude that ET-1 decreases PPARγ signaling and contributes to PAEC dysfunction and impaired angiogenesis in PPHN. We speculate that therapies aimed at decreasing ET-1 production will restore PPARγ signaling, preserve endothelial function, and improve angiogenesis in PPHN.


Asunto(s)
Células Endoteliales/metabolismo , Endotelina-1/fisiología , Neovascularización Fisiológica , PPAR gamma/metabolismo , Animales , Bosentán , Células Cultivadas , Antagonistas de los Receptores de Endotelina , Humanos , Recién Nacido , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , PPAR gamma/agonistas , Síndrome de Circulación Fetal Persistente/metabolismo , Síndrome de Circulación Fetal Persistente/fisiopatología , Arteria Pulmonar/patología , Receptores de Endotelina/metabolismo , Ovinos , Transducción de Señal , Sulfonamidas/farmacología
16.
J Hazard Mater ; 471: 134337, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38640674

RESUMEN

BACKGROUND: Hexafluoropropylene oxide trimer acid (HFPO-TA), a perfluorooctanoic acid (PFOA) substitute, exhibited strong affinity and capability to activate peroxisome proliferator activated receptor gamma (PPARγ), a lipid metabolism regulator, suggesting potential to induce metabolic toxicities. METHODS: Fertile chicken eggs were exposed to 0, 0.5, 1 or 2 mg/kg (egg weight) HFPO-TA and incubated until hatch. Serum from 0- and 3- month-old chickens were subjected to liquid chromatography ultra-high resolution mass spectrometry for HFPO-TA concentration, while liver, pancreas and adipose tissue samples were collected for histopathological assessments. In ovo PPARγ reporter and silencing system were established with lentivirus microinjection. qRT-PCR and immunohistochemistry were utilized to evaluate the expression levels of PPARγ downstream genes. RESULTS: In 3-month-old animals developmentally exposed to HFPO-TA, adipose tissue hyperplasia, hepatic steatosis, pancreas islet hypertrophy and elevated serum free fatty acid / insulin levels were observed. Results of reporter assay and qRT-PCR indicated HFPO-TA-mediated PPARγ transactivation in chicken embryo. Silencing of PPARγ alleviated HFPO-TA-induced changes, while PPARγ agonist rosiglitazone mimicked HFPO-TA-induced effects. qRT-PCR and immunohistochemistry revealed that FASN and GPD1 were upregulated following developmental exposure to HFPO-TA in 3-month-old animals. CONCLUSIONS: Developmental exposure to HFPO-TA induced persistent metabolic toxicities in chickens, in which PPARγ played a central role.


Asunto(s)
Fluorocarburos , PPAR gamma , Animales , PPAR gamma/genética , PPAR gamma/metabolismo , Fluorocarburos/toxicidad , Embrión de Pollo , Hígado/efectos de los fármacos , Hígado/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Pollos , Páncreas/efectos de los fármacos , Páncreas/metabolismo
17.
Chin J Cancer Res ; 25(6): 704-14, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24385698

RESUMEN

OBJECTIVE: To explore the relationship between peroxisome proliferator activated receptor-gamma (PPARγ) and peroxisome proliferator-activated receptor-gamma coactivator-1 (PGC-1) expression in gastric carcinoma (GC), and analyze their correlations with clinicopathological features and clinical outcomes of patients. METHODS: The two-step immunohistochemical method was used to detect the expression of PPARγ and PGC-1 in 179 cases of GC, and 108 cases of matched normal gastric mucosa. Besides, 16 cases of fresh GC specimens and corresponding normal gastric mucosa were detected for PGC-1 expression with Western blotting. RESULTS: The positive rates of PPARγ and PGC-1 expression were significantly lower in GC (54.75%, 49.16%) than in normal gastric mucosa (70.37%, 71.30%), respectively (P<0.05). The decreased expression of PGC-1 in GC was confirmed in our Western blot analysis (P=0.004). PPARγ and PGC-1 expressions were related to Lauren's types of GC (P<0.05). Positive correlation was found between PPARγ and PGC-1 expression in GC (rk=0.422, P<0.001). The survival time of PPARγ negative and positive patients was 36.6±3.0 vs. 38.5±2.7 months, and no statistical difference was found between the 5-year survival rates of two groups (34.4% vs. 44.1%, P=0.522, log-rank test); the survival time of PGC-1 negative and positive patients was 36.2±2.8 vs. 39.9±2.9 months, while no statistical difference was found between the 5-year survival rates of the two groups (32.0% vs. 48.2%, P=0.462, log-rank test). CONCLUSIONS: Decreased expression of PPARγ and PGC-1 in GC was related to the Lauren's classification. Their expressions in GC were positively correlated, indicating that their functions in gastric carcinogenesis may be closely related.

18.
Artículo en Inglés | MEDLINE | ID: mdl-36535597

RESUMEN

Total absence of adipose tissue (lipoatrophy) is associated with the development of severe metabolic disorders including hepatomegaly and fatty liver. Here, we sought to investigate the impact of severe lipoatrophy induced by deletion of peroxisome proliferator-activated receptor gamma (PPARγ) exclusively in adipocytes on lipid metabolism in mice. Untargeted lipidomics of plasma, gastrocnemius and liver uncovered a systemic depletion of the essential linoleic (LA) and α-linolenic (ALA) fatty acids from several lipid classes (storage lipids, glycerophospholipids, free fatty acids) in lipoatrophic mice. Our data revealed that such essential fatty acid depletion was linked to increased: 1) capacity for liver mitochondrial fatty acid ß-oxidation (FAO), 2) citrate synthase activity and coenzyme Q content in the liver, 3) whole-body oxygen consumption and reduced respiratory exchange rate in the dark period, and 4) de novo lipogenesis and carbon flux in the TCA cycle. The key role of de novo lipogenesis in hepatic steatosis was evidenced by an accumulation of stearic, oleic, sapienic and mead acids in liver. Our results thus indicate that the simultaneous activation of the antagonic processes FAO and de novo lipogenesis in liver may create a futile metabolic cycle leading to a preferential depletion of LA and ALA. Noteworthy, this previously unrecognized cycle may also explain the increased energy expenditure displayed by lipoatrophic mice, adding a new piece to the metabolic regulation puzzle in lipoatrophies.


Asunto(s)
Hígado Graso , Lipogénesis , Animales , Ratones , Ciclo del Sustrato , Metabolismo de los Lípidos , Hígado Graso/metabolismo , Ácido alfa-Linolénico/metabolismo
19.
Environ Int ; 173: 107857, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36881956

RESUMEN

Perfluoroalkyl substances (PFAS) are persistent and pose a risk to human health. High throughput screening (HTS) cell-based bioassays may inform risk assessment of PFAS provided that quantitative in vitro to in vivo extrapolation (QIVIVE) can be developed. The QIVIVE ratio is the ratio of nominal (Cnom) or freely dissolved concentration (Cfree) in human blood to Cnom or Cfree in the bioassays. Considering that the concentrations of PFAS in human plasma and in vitro bioassays may vary by orders of magnitude, we tested the hypothesis that anionic PFAS bind to proteins concentration-dependently and therefore the binding differs substantially between human plasma and bioassays, which has an impact on QIVIVE. Solid phase microextraction (SPME) with C18-coated fibers served to quantify the Cfree of four anionic PFAS (perfluorobutanoate (PFBA), perfluorooctanoate (PFOA), perfluorohexane sulfonate (PFHxS) and perfluorooctane sulfonate (PFOS)) in the presence of proteins and lipid, medium components, cells and human plasma over five orders of magnitude in concentrations. The C18-SPME method was used to quantify the non-linear binding to proteins, human plasma and medium, and the partition constants to cells. These binding parameters were used to predict Cfree of PFAS in cell bioassays and human plasma by a concentration-dependent mass balance model (MBM). The approach was illustrated with a reporter gene assay indicating activation of the peroxisome proliferator-activated receptor gamma (PPARγ-GeneBLAzer). Blood plasma levels were collected from literature for occupational exposure and the general population. The QIVIVEnom ratios were higher than the QIVIVEfree ratios due to the strong affinity to proteins and large differences in protein contents between human blood and bioassays. For human health risk assessment, the QIVIVEfree ratios of many in vitro assays need to be combined to cover all health relevant endpoints. If Cfree cannot be measured, they can be estimated with the MBM and concentration-dependent distribution ratios.


Asunto(s)
Ácidos Alcanesulfónicos , Contaminantes Ambientales , Fluorocarburos , Humanos , Disponibilidad Biológica , Unión Proteica , Fluorocarburos/toxicidad , Ácidos Alcanesulfónicos/toxicidad , Alcanosulfonatos , Bioensayo
20.
Mol Neurobiol ; 59(7): 4368-4383, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35553009

RESUMEN

Telmisartan (TEL) is an angiotensin II type 1 receptor blocker and a partial activator of peroxisome proliferator-activated receptor-gamma (PPARγ), which regulates inflammatory and apoptotic pathways. Increasing evidence has demonstrated the PPARγ agonistic property of TEL in several brain disorders. This study aims to explore the neuroprotective impact of TEL in 3-nitropropionic acid (3-NP)-induced neurotoxicity in rats. The PPARγ effect of TEL was affirmed by using the PPARγ agonist pioglitazone (PIO), and the antagonist GW9662. 3-NP led to a significant reduction in body weight alongside motor and cognitive functioning. The striata of the 3-NP-treated rats showed energy-deficit, microglia-mediated inflammatory reactions, apoptotic damage as well as histopathological lesions. PIO and TEL improved motor and cognitive perturbations induced by 3-NP, as confirmed by striatal histopathological examination, energy restoration, and neuronal preservation. Both drugs improved mitochondrial biogenesis evidenced by elevated mRNA expression of PPARγ, PGC-1α, and TFAM, alongside increased striatal ATP and SDH. The mitochondrial effect of TEL was beyond PPARγ activation. As well, their anti-inflammatory effect was attributed to suppression of microglial activation, and protein expression of pS536 p65 NF-κB with marked attenuation of striatal inflammatory mediator's release. Anti-inflammatory cytokine IL-10 expression was concurrently increased. TEL effectively participated in neuronal survival as it promoted phosphorylation of Akt/GSK-3ß, further increased Bcl-2 expression, and inhibited cleavage of caspase-3. Interestingly, co-treatment with GW9662 partially revoked the beneficial effects of TEL. These findings recommend that TEL improves motor and cognitive performance, while reducing neuronal inflammation and apoptosis in 3-NP-induced neurotoxicity via a PPARγ-dependent mechanism.


Asunto(s)
PPAR gamma , Propionatos , Animales , Glucógeno Sintasa Quinasa 3 beta , Nitrocompuestos , PPAR gamma/metabolismo , Pioglitazona/farmacología , Propionatos/toxicidad , Ratas , Telmisartán/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA