Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Zhongguo Zhong Yao Za Zhi ; 47(18): 5022-5031, 2022 Sep.
Article in Chinese | MEDLINE | ID: mdl-36164912

ABSTRACT

The saponins in different parts of Gynostemma pentaphyllum were analyzed via UPLC-Q-TOF-MS~E. A total of 46 saponins were identified, and the underground part had 26 saponins more than the aboveground part, most of which were trisaccharide saponins. The rat model of hyperlipidemia was established with high-fat diet. This study explored the lipid-lowering activity of total saponins in the underground part of G. pentaphyllum, so as to provide a theoretical basis for the comprehensive utilization of the underground part of G. pentaphyllum. A total of 99 healthy SD rats were randomly assigned into a blank group, a model group, a positive drug group, an aboveground total saponins group, and low-, medium-, and high-dose underground total saponins groups. Except the blank group, the other groups were fed with high-fat diet for 6 weeks. Then, the blood was collected from the orbital cavity to determine whether the modeling was successful according to the serum levels of total cholesterol(TC) and triglyceride(TG). After intragastric administration of the corresponding agents for 30 continuous days, the physical state of the rats were observed, and the body weight and liver specific gravity were measured. Furthermore, the levels of TC, TG, low-density lipoprotein cholesterol(LDL-C), high-density lipoprotein cholesterol(HDL-C), alanine transaminase(ALT), aspartate transaminase(AST), bilirubin, and total bile acids in serum, as well as the levels of superoxide dismutase(SOD), malondialdehyde(MDA), peroxidase proliferator-activated receptor(PPAR-γ) in the liver tissue, were determined. The pathological changes of liver was observed via HE staining. The results showed that the aboveground total saponins and medium-and high-dose underground total saponins can treat hepatocyte steatosis, lower TC, TG, LDL-C, ALT, AST, total bilirubin, MDA, and PPAR-γ levels, and increase HDL-C and SOD levels in the model rats. The effect tended to be more obvious with the increase in dosage. Therefore, the total saponins in the underground part of G. pentaphyllum have good pharmacological effect of reducing blood lipid, which provides a theoretical basis for the comprehensive utilization of the underground part of G. pentaphyllum.


Subject(s)
Gynostemma , Hypolipidemic Agents , Saponins , Alanine Transaminase/analysis , Animals , Aspartate Aminotransferases/analysis , Bile Acids and Salts/blood , Bilirubin/blood , Cholesterol, LDL/blood , Diet, High-Fat/adverse effects , Gynostemma/chemistry , Hypolipidemic Agents/pharmacology , Hypolipidemic Agents/therapeutic use , Lipoproteins, HDL/blood , Liver/chemistry , Liver/metabolism , Malondialdehyde/analysis , Peroxisome Proliferator-Activated Receptors/analysis , Rats , Rats, Sprague-Dawley , Saponins/pharmacology , Saponins/therapeutic use , Superoxide Dismutase , Triglycerides/blood , Trisaccharides/pharmacology , Trisaccharides/therapeutic use
2.
Kidney Blood Press Res ; 42(3): 398-405, 2017.
Article in English | MEDLINE | ID: mdl-28668962

ABSTRACT

BACKGROUND/AIMS: Carnitine is essential for the transport of long-chain FAs (FA) into the mitochondria for energy production. During acute exercise, the increased demand for FAs results in a state of free carnitine deficiency in plasma. The role of kidney in carnitine homeostasis after exercise is not known. METHODS: Swiss Webster mice were sacrificed immediately after a 1-hour moderate intensity treadmill run, and at 4-hours and 8-hours into recovery. Non-exercising mice served as controls. Plasma was analyzed for carnitine using acetyltransferase and [14C] acetyl-CoA. Kidney was removed for gene and protein expression of butyrobetaine hydroxylase (γ-BBH), organic cation transporter (OCTN2), and peroxisome proliferator-activated receptor (PPARα), a regulator of fatty acid oxidation activated by FAs. RESULTS: Acute exercise caused a decrease in plasma free carnitine levels. Rapid return of free carnitine to control levels during recovery was associated with increased γ-BBH expression. Both mRNA and protein levels of OCTN2 were detected in kidney after exercise and during recovery, suggesting renal transport mechanisms were stimulated. These changes were accompanied with a reciprocal increase in PPARα protein expression. CONCLUSIONS: Our results show that the decrease in free carnitine after exercise rapidly activates carnitine biosynthesis and renal transport mechanism in kidney to establish carnitine homeostasis.


Subject(s)
Carnitine/biosynthesis , Kidney/metabolism , Physical Conditioning, Animal , Solute Carrier Family 22 Member 5/metabolism , Animals , Carnitine/blood , Fatty Acids , Homeostasis , Mice , Peroxisome Proliferator-Activated Receptors/analysis , Physical Conditioning, Animal/physiology , Solute Carrier Family 22 Member 5/analysis
3.
Theriogenology ; 101: 53-61, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28708516

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor family of ligand-dependent transcription factors. PPARs are critical regulators of glucose homeostasis and lipid metabolism, and affect cell proliferation and differentiation. In the current study, we examined (1) the profiles of PPARA, PPARD, and PPARG mRNA expression and DNA binding activity in porcine conceptuses collected on Days 10-11 (spherical and tubular conceptuses), 11-12 (filamentous conceptuses), 13-14, and 15-16 (elongated conceptuses) of pregnancy, (2) the presence of PPARA, PPARD, and PPARG proteins in Days 10, 12, and 15 conceptuses. Moreover, we analyzed the abundance of retinoid X receptor (RXR; PPARs heterodimer partner) transcripts as well as the correlation between PPARs mRNA expression and the expression of genes important for and/or associated with elongation of porcine conceptuses: aromatase (CYP19A1), prostaglandin endoperoxide synthase 2 (PTGS2), glucose transporter 1 (SLC2A1), and interleukin 1B (IL1B). PPARA mRNA expression in conceptuses did not change during Days 10-14 of gestation, but was greater on Days 15-16 compared to Days 10-11 (P < 0.05). A considerable increase in PPARD and PPARG mRNA expression was observed in filamentous conceptuses from Days 11-12 compared to spherical and tubular conceptuses from Days 10-11 (P < 0.01), followed by a decrease on Days 13-14 and 15-16 (P < 0.05). PPARA, PPARD, and PPARG proteins were present in conceptus tissue demonstrating nuclear localization clearly visible on Days 12 and 15 of pregnancy. DNA binding activity of the PPARD isoform was greater in filamentous conceptuses from Days 11-12 than in spherical and tubular conceptuses from Days 10-11 (P < 0.01). Moreover, concentrations of active PPARD and PPARG proteins in nuclear fractions of conceptus tissue were greater on Days 11-12 compared to Days 13-14 and 15-16 of pregnancy (P < 0.05). RXRA, RXRD, and RXRG mRNA expression in conceptuses increased on Days 11-12 compared to Days 10-11 (P < 0.05). PPARD and PPARG mRNA expression showed strong positive correlations with PTGS2 mRNA expression (P < 0.0001). Additionally, PPARD gene expression correlated with SLC2A1 and IL1B mRNA expression (P < 0.01). Collectively, these results indicate that among all three PPARs expressed in peri-implantation porcine conceptuses, PPARD and PPARG may be involved in conceptus elongation before implantation.


Subject(s)
Blastocyst/chemistry , Peroxisome Proliferator-Activated Receptors/analysis , Protein Isoforms/analysis , Sus scrofa/embryology , Animals , DNA/metabolism , Embryo Implantation , Female , Gene Expression , PPAR alpha/genetics , PPAR delta/genetics , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Pregnancy
4.
Nutr. hosp ; 33(6): 1354-1360, nov.-dic. 2016. tab
Article in English | IBECS | ID: ibc-159815

ABSTRACT

Introduction: Breast cancer is the second most common cancer in the world, and the most frequent cancer among women. Moreover, there are factors that influence the risk for breast cancer including the age, genetic and endocrine factors, and lifestyle. Objectives: To evaluate the consumption of fatty acids; compare the fatty acids composition in the breast adipose tissue of women with breast cancer and benign breast disease as well as potential risk factors; and describe the genotypic frequency of the Pro12Ala PPARγ polymorphism. Material and methods: A hospital-based case-control study was conducted including incident cases (n = 38 breast cancer; n = 75 benign breast disease; n = 166 control). Lifestyle features, socioeconomic issues, dietary intake, anthropometry, and blood and tissue data were assessed. Results: No differences were observed for fatty acids intake. Interestingly, lauric acid (p = 0.001), myristic acid (p = 0.036), stearic acid (p = 0.031), and total saturated fatty acids (SFAs) (p = 0.048) had lower concentrations in BC than in BBD women, while palmitoleic acid (p = 0.022), erucic acid (p = 0.002), total monounsaturated fatty acids (MUFAs) (p = 0.039) and oleic acid/stearic acid ratio (p = 0.015) increased. There was no significant association between PPARγ polymorphism and studied groups (p = 0.977). The age at first full pregnancy (p = 0.004) was significantly associated with the development BC, whereas BMI (p = 0.005); percentage of body fat (p = 0.024); physical activity (p = 0.036); and age at menarche (p = 0.008), at first full pregnancy (p < 0.001), and of first mammogram (p = 0.018) were significantly associated with the development of BBD. Conclusion: The results suggest a different fatty acids composition of breast adipose tissue, a biomarker of long-term dietary intake, particularly for SFAs, MUFA and 18: 1 n-9/18: 00 ratio. Our findings also show that are differences in the factors related to the development of BC and BBC (AU)


Introducción: el cáncer de mama (CM) es el segundo cáncer más común en el mundo, y el cáncer más frecuente entre las mujeres. Por otra parte, hay factores que influyen en el riesgo de padecer CM, entre los que se encuentran la edad, factores genéticos y endocrinos, y el estilo de vida. Objetivos: evaluar el consumo de ácidos grasos; comparar la composición de ácidos grasos en el tejido adiposo de mama de las mujeres con CM y enfermedad benigna de mama (EBM), así como los posibles factores de riesgo; y describir la frecuencia genotípica del polimorfismo Pro12Ala PPARγ. Material y métodos: se llevó a cabo un estudio caso-control basado en hospitales, incluyendo casos incidentes (n = 38 cáncer de mama, n = 75 enfermedad benigna de mama, n = 166 control). Se evaluaron las características del estilo de vida, las cuestiones socioeconómicas, la ingesta dietética, la antropometría y los datos de sangre y tejidos. Resultados: no se observaron diferencias para la ingesta de ácidos grasos. Curiosamente, ácido láurico (p = 0,001), ácido mirístico (p = 0,036), ácido esteárico (p = 0,031) y los ácidos grasos totales saturados (AGS) (p = 0,048) tenían concentraciones más bajas en CM que en mujeres EBM, mientras ácido palmitoleico (p = 0,022), ácido erúcico (p = 0,002), los ácidos totales grasos monoinsaturados (MUFA) (p = 0,039) y la relación ácido oleico/ácido esteárico (p = 0,015) aumentó. No hubo asociación significativa entre el polimorfismo PPAR gamma y los grupos de estudio (p = 0,977). La edad al primer embarazo (p = 0,004) se asoció de forma signifi cativa con el desarrollo de CM, mientras que el IMC (p = 0,005), porcentaje de grasa corporal (p = 0,024), la actividad física (p = 0,036) y la edad de la menarquia (p = 0,008), al primer embarazo (p < 0,001), y de la primera mamografía (p = 0,018), fueron significativamente asociados con el desarrollo de EBM. Conclusiones: los resultados sugieren una composición diferente de ácidos grasos del tejido adiposo de la mama, un biomarcador de la ingesta dietética a largo plazo, particularmente para SFA, MUFA y 18: 1 n-9/18: 00. Nuestros hallazgos también muestran que existen diferencias en los factores relacionados con el desarrollo de CM y EBM (AU)


Subject(s)
Humans , Female , Breast Neoplasms/pathology , Fibrocystic Breast Disease/pathology , Adipose Tissue/pathology , Fatty Acids/analysis , Biomarkers, Tumor/analysis , Polymorphism, Genetic , Dietary Fats/analysis , Peroxisome Proliferator-Activated Receptors/analysis
5.
An. R. Acad. Farm ; 82(n.extr): 206-214, oct. 2016. tab
Article in Spanish | IBECS | ID: ibc-157626

ABSTRACT

La incidencia y prevalencia de sobrepeso y obesidad ha experimentado un gran incremento en las últimas décadas. Aunque el balance entre ingreso y gasto energético es clave en la inducción y establecimiento de la obesidad, otros factores parecen estar netamente implicados. A este respecto la hipótesis de los obesógenos ambientales, ha ganado credibilidad en los años recientes, al identificarse agentes químicos que promueven la adipogénesis y obesidad en animales y humanos. En esta revisión, se hace un estudio de los disruptores endocrinos que son sustancias químicas contaminantes del medio ambiente y su relación con la obesidad. Se revisan los principales tipos de obesógenos, p.ej. dietilestilbestrol, bisfenol A, los compuestos orgánicos derivados del estaño, la genisteína y los ftalatos y sus características diferenciales y mecanismos de acción. Se ultima con unas reflexiones sobre la necesidad de profundizar en el conocimiento de estos compuestos tan abundantes hoy en día (AU)


Overweight and obesity incidence and prevalence have increased during the last decades. Although the energy intake/expenditure balance is a key factor in the obesity induction and development, other factors seem clearly implicated. In fact the environmental obesogens hypothesis has gain importance in the last years, as different chemical agents have been found to adipogenics and obesogenics in animal models and humans. In this review we study the role of environmental chemical polluting as endocrine disruptors and their relationship with obesity. Major obesogen types, e.g. diethylstilboestrol, bisphenol A, tinorganic derived compounds, genistein and ftalates are described. Their differential characteristics and action mechanisms are defined. The review ends with some considerations on the need to get knowledge on such abundant compounds (AU)


Subject(s)
Humans , Obesity/etiology , Environmental Pollutants/adverse effects , Endocrine Disruptors/analysis , Adipogenesis/physiology , Gene-Environment Interaction , Epigenesis, Genetic , Peroxisome Proliferator-Activated Receptors/analysis , Diethylhexyl Phthalate/analysis , Reactive Oxygen Species/analysis , Organic Chemicals/analysis
6.
Sci Rep ; 6: 30051, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27444220

ABSTRACT

Energy metabolism, involving the ATP-dependent AMPK-PgC-Ppar pathway impacts metabolic health immensely, in that its impairment can lead to obesity, giving rise to disease. Based on observations that individuals with Gilbert's syndrome (GS; UGT1A1(*)28 promoter mutation) are generally lighter, leaner and healthier than controls, specific inter-group differences in the AMPK pathway regulation were explored. Therefore, a case-control study involving 120 fasted, healthy, age- and gender matched subjects with/without GS, was conducted. By utilising intra-cellular flow cytometry (next to assessing AMPKα1 gene expression), levels of functioning proteins (phospho-AMPK α1/α2, PgC 1 α, Ppar α and γ) were measured in PBMCs (peripheral blood mononucleated cells). In GS individuals, rates of phospho-AMPK α1/α2, -Ppar α/γ and of PgC 1α were significantly higher, attesting to a boosted fasting response in this condition. In line with this finding, AMPKα1 gene expression was equal between the groups, possibly stressing the post-translational importance of boosted fasting effects in GS. In reflection of an apparently improved health status, GS individuals had significantly lower BMI, glucose, insulin, C-peptide and triglyceride levels. Herewith, we propose a new theory to explain why individuals having GS are leaner and healthier, and are therefore less likely to contract metabolic diseases or die prematurely thereof.


Subject(s)
AMP-Activated Protein Kinases/analysis , Gilbert Disease/pathology , Leukocytes, Mononuclear/enzymology , Metabolic Networks and Pathways , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/analysis , Peroxisome Proliferator-Activated Receptors/analysis , AMP-Activated Protein Kinases/genetics , Adolescent , Adult , Case-Control Studies , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptors/genetics , Young Adult
7.
Histol Histopathol ; 31(9): 1029-36, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26867926

ABSTRACT

INTRODUCTION: PPAR expression in placenta tissues regulates proinflammatory cytokine production and preserves the quiescence of the uterus during pregnancy. PPAR-γ regulates inflammatory response during gestation while PPAR-δ and TNFα play a central role at implantation, decidualization and placentation. However, their expression levels affect normal pregnancy and may cause gestational complications and miscarriage. The aim of this report is to investigate the relationship of these molecules with unexplained recurrent miscarriage. MATERIALS-METHODS: The miscarriage group was obtained from 12 women, between the ages of 35 to 42 years, who miscarried during the 1st trimester of gestation and controls consisted of 12 healthy women, between the ages of 27 to 39 years, who had electively terminated their pregnancies, during the 1st trimester of gestation. The abortion material was processed and specimens taken were studied using immunohisto-chemical methods. Specimens were taken from decidua basalis and decidua parietalis. Monoclonal antibodies were used against PPAR-γ (Peroxisome Proliferator Activation Receptor γ), PPAR-δ and TNFα (Tumor Necrosis Factor alpha). The results were statistically analyzed with Mann-Whitney test. RESULTS: Our research identified PPAR-γ expression in decidua basalis and decidua parietalis from control group and decidua basalis from miscarriage group. PPAR-δ expression was also identified in both deciduas from both groups. Statistically, no significant change in PPAR-γ and PPAR-δ expression was observed between recurrent miscarriage group and controls. On the contrary, a statistically significant upregulation of TNFα was identified in both deciduas between miscarriage group and controls (p<0.05). CONCLUSIONS: Our evidence did not support a possible role of PPARs expression in recurrent pregnancy loss. However, a potential involvement of TNFα in the syndrome was reported. Further research should be performed due to insufficient bibliographic data.


Subject(s)
Abortion, Habitual/metabolism , Peroxisome Proliferator-Activated Receptors/biosynthesis , Placenta/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Adult , Female , Humans , Immunohistochemistry , Peroxisome Proliferator-Activated Receptors/analysis , Pregnancy , Pregnancy Trimester, First , Tumor Necrosis Factor-alpha/analysis
8.
Endokrynol Pol ; 66(4): 334-41, 2015.
Article in English | MEDLINE | ID: mdl-26323471

ABSTRACT

INTRODUCTION: A high-altitude environment causes appetite loss in unacclimatised humans, leading to weight reduction. Ghrelin, cholecystokinin (CCK), and glucagon like peptide-1 (GLP-1), are gut hormones involved in the regulation of food intake and energy metabolism. The liver is an important site of metabolic regulation, and together with the gut it plays a role in food intake regulation. This study intends to study the time-dependent changes occurring in plasma gut hormones, PPARα, PPARδ, and PGC1α, in the stomach and liver during hypoxia. MATERIAL AND METHODS: Male Sprague Dawley rats were exposed to hypobaric hypoxia in a decompression chamber at 7620 m for different durations up to seven days. RESULTS: Hypoxia increased circulating ghrelin from the third day onwards while CCK and GLP-1 decreased immediately. An increase in ghrelin, ghrelin receptor protein levels, and GOAT mRNA levels in the stomach was observed. Stomach cholecystokinin receptor (CCKAR), PPARα, and PPARδ decreased. Liver CCKAR decreased during the first day of hypoxia and returned to normal levels from the third day onwards. PPARα and PGC1α expression increased while PPARδ protein levels reduced in the liver on third day. CONCLUSION: Hypoxia alters the expression of ghrelin and ghrelin receptor in the stomach, CCKAR in the liver, and PPAR and its cofactors, which might be possible role players in the contribution of gut and liver to anorexia at high altitude.


Subject(s)
Anorexia/etiology , Cholecystokinin/genetics , Disease Models, Animal , Ghrelin/genetics , Glucagon-Like Peptide 1/genetics , Hypoxia/complications , Peroxisome Proliferator-Activated Receptors/genetics , Animals , Anorexia/metabolism , Cholecystokinin/analysis , Gastric Mucosa/metabolism , Gene Expression Regulation , Ghrelin/analysis , Glucagon-Like Peptide 1/analysis , Hypoxia/metabolism , Liver/metabolism , Male , Peroxisome Proliferator-Activated Receptors/analysis , Rats , Rats, Sprague-Dawley
9.
Toxicology ; 336: 84-95, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26219505

ABSTRACT

Triptolide is the major active ingredient of Tripterygium Glycosides (TG), a traditional Chinese medicine with very potent anti-inflammatory effects and has been used in China for the treatment of rheumatoid arthritis and many other inflammatory diseases. However, clinical application of triptolide is restricted due to its multiple side effects, especially male infertility. The mechanism of triptolide on reproduction toxicity remains unclear. In the present study, a GC-MS based metabolomic approach was employed to evaluate the mechanism of triptolide-induced reproductive toxicity as well as identify potential novel biomarkers for the early detection of spermatogenesis dysfunction. In brief, male mice were divided into two groups with or without triptolide intraperitoneal injection at 60 µg/kg/day for 2 weeks and toxic effect of triptolide on testicular tissues were examined by biochemical indicator analysis, testis histopathologic analysis, and sperm quantity analysis. Metabolomics technology was then performed to evaluate systematically the endogenous metabolites profiling. Our results demonstrated that triptolide suppressed the marker-enzymes of spermatogenesis and testosterone levels, decreased sperm counts, reduced the gonad index and destroyed the microstructure of testis. Multivariate data analysis revealed that mice with triptolide induced testicular toxicity could be distinctively differentiated from normal animals and 35 and 39 small molecule metabolites were changed significantly in testis and serum, respectively (Fold-changes >1.5, P<0.05), in triptolide-treated mice. Abnormal level of fatty acids, an important energy source of sertoli cells with critical role in maintaining normal function of the testis tissue, was observed in triptolide-treated mice. Additionally, the protein expressions of PPAR, a transcription factor known to play a pivotal role in lipid and energy metabolism was significantly decreased in the testis tissue of triptolide-treated mice. In summary, our study represents the first comprehensive GC-MS based metabolomics analysis of triptolide-induced testicular toxicity. We reported for the first time that exposure to triptolide led to marked changes of a panel of endogenous metabolites in both testis and serum. The impairment of spermatogenesis may be caused by abnormal lipid and energy metabolism in testis via the down-regulation of PPARs mediated by triptolide. The presence of research suggested that PPARs and its related fatty acids metabolism may serve as potential targets for intervention or treatment of male infertility induced by triptolide.


Subject(s)
Anti-Inflammatory Agents/toxicity , Diterpenes/toxicity , Fatty Acids/analysis , Peroxisome Proliferator-Activated Receptors/analysis , Phenanthrenes/toxicity , Testis/drug effects , Acetylcarnitine/analysis , Animals , Carnitine/analysis , Epoxy Compounds/toxicity , Gas Chromatography-Mass Spectrometry , Male , Metabolomics , Mice , Spermatogenesis/drug effects , Spermatozoa/drug effects , Testis/chemistry , Testis/metabolism , Testosterone/blood
10.
J Endod ; 40(8): 1132-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25069920

ABSTRACT

INTRODUCTION: Dental pulp has limited capability to regenerate, which happens in the early stage of pulpitis. An ambiguous relationship exists; inflammation may impair or support pulp regeneration. Epigenetics, which is involved in cell proliferation and inflammation, could regulate human dental pulp cell (HDPCs) regeneration. The aim of this study was to determine the role of the epigenetic mark, enhancer of zeste homolog 2 (EZH2), in the inflammation, proliferation, and regeneration of dental pulp. We used trimethylated histone H3 lysine 27(H3K27me3) and its lysine demethylase 6B (KDM6B) to monitor functional effects of altered EZH2 levels. METHODS: We detected epigenetic marks (EZH2, H3K27me3, and KDM6B) in pulp tissue by immunohistochemistry and immunofluorescence. EZH2 levels in HDPCs in inflammatory responses or differentiation were analyzed by quantitative polymerase chain reaction and Western blot. Quantitative polymerase chain reaction was used to assess the effects of EZH2 inhibition on interleukins in HDPCs upon tumor necrosis factor alpha stimulation. Cell proliferation was tested by cell counting kit-8, cell cycle, and apoptosis analysis. HDPC differentiation was investigated by quantitative polymerase chain reaction, alkaline phosphatase activity, and oil red O staining. RESULTS: EZH2 and H3K27me3 were decreased, whereas KDM6B was increased in infected pulp tissue and cells, which were similar to HDPC differentiation. EZH2 inhibition suppressed IL-1b, IL-6, and IL-8 messenger RNA (mRNA) in HDPCs upon inflammatory stimuli and impeded HDPC proliferation by decreasing cell number, arresting cell cycle, and increasing apoptosis. Suppressed EZH2 impaired adipogenesis, peroxisome proliferator-activated receptor r (PPAR-r), and CCAAT-enhancer binding protein a (CEBP/a) mRNA in adipogenic induction while enhancing alkaline phosphatase activity, Osx, and bone sialoprotein (BSP) mRNA in mineralization induction of HDPCs. CONCLUSIONS: EZH2 inhibited HDPC osteogenic differentiation while enhancing inflammatory response and proliferation, suggesting its role in pulp inflammation, proliferation, and regeneration.


Subject(s)
Dental Pulp/physiology , Polycomb Repressive Complex 2/physiology , Pulpitis/physiopathology , Regeneration/physiology , Adipogenesis/physiology , Alkaline Phosphatase/analysis , Apoptosis/physiology , CCAAT-Enhancer-Binding Protein-alpha/analysis , Calcification, Physiologic/physiology , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Dental Pulp/cytology , Dental Pulp/drug effects , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic/physiology , Histones/analysis , Humans , Integrin-Binding Sialoprotein/analysis , Interleukin-1beta/analysis , Interleukin-6/analysis , Interleukin-8/analysis , Jumonji Domain-Containing Histone Demethylases/analysis , Osteogenesis/physiology , Peroxisome Proliferator-Activated Receptors/analysis , Polycomb Repressive Complex 2/antagonists & inhibitors , Sp7 Transcription Factor , Transcription Factors/analysis , Tumor Necrosis Factor-alpha/pharmacology
11.
J Comp Pathol ; 149(1): 10-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23219070

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors belonging to the nuclear hormone receptor superfamily. Recent studies have demonstrated that PPARs regulate lipid metabolism and are expressed in various cancers. The aim of the present study was to investigate the expression of PPAR-α, -ß and -γ in normal canine testicular tissue and canine testicular tumours (CTTs). Expression of PPAR-α, -ß and -γ was greater (P <0.05) than in normal testicular tissue. PPARs were therefore induced in CTTs and they may play a role in the biology of these tumours.


Subject(s)
Dog Diseases/metabolism , Peroxisome Proliferator-Activated Receptors/biosynthesis , Testicular Neoplasms/veterinary , Testis/metabolism , Animals , Dogs , Immunohistochemistry , Male , Peroxisome Proliferator-Activated Receptors/analysis , Testicular Neoplasms/metabolism
12.
PLoS One ; 7(4): e34969, 2012.
Article in English | MEDLINE | ID: mdl-22496881

ABSTRACT

Chitosan has been widely used in food industry as a weight-loss aid and a cholesterol-lowering agent. Previous studies have shown that chitosan affects metabolic responses and contributes to anti-diabetic, hypocholesteremic, and blood glucose-lowering effects; however, the in vivo targeting sites and mechanisms of chitosan remain to be clarified. In this study, we constructed transgenic mice, which carried the luciferase genes driven by peroxisome proliferator-activated receptor (PPAR), a key regulator of fatty acid and glucose metabolism. Bioluminescent imaging of PPAR transgenic mice was applied to report the organs that chitosan acted on, and gene expression profiles of chitosan-targeted organs were further analyzed to elucidate the mechanisms of chitosan. Bioluminescent imaging showed that constitutive PPAR activities were detected in brain and gastrointestinal tract. Administration of chitosan significantly activated the PPAR activities in brain and stomach. Microarray analysis of brain and stomach showed that several pathways involved in lipid and glucose metabolism were regulated by chitosan. Moreover, the expression levels of metabolism-associated genes like apolipoprotein B (apoB) and ghrelin genes were down-regulated by chitosan. In conclusion, these findings suggested the feasibility of PPAR bioluminescent imaging-guided transcriptomic analysis on the evaluation of chitosan-affected metabolic responses in vivo. Moreover, we newly identified that downregulated expression of apoB and ghrelin genes were novel mechanisms for chitosan-affected metabolic responses in vivo.


Subject(s)
Anticholesteremic Agents/pharmacology , Chitosan/pharmacology , Gene Expression Profiling , Luminescent Measurements , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Apolipoproteins B/biosynthesis , Brain/metabolism , Down-Regulation/drug effects , Gastrointestinal Tract/metabolism , Ghrelin/biosynthesis , Glucose/metabolism , Lipid Metabolism/drug effects , Male , Mice , Mice, Transgenic , Peroxisome Proliferator-Activated Receptors/analysis
13.
Clín. investig. arterioscler. (Ed. impr.) ; 24(2): 89-91, mar.-abr. 2012.
Article in Spanish | IBECS | ID: ibc-105079

ABSTRACT

Introducción La suplementación de la dieta con un 10% (p/v) de fructosa en el agua de bebida durante 14 días en ratas produce hipertrigliceridemia y esteatosis hepática como consecuencia de una reducción en la expresión y en la actividad transcripcional de (..) (AU)


Introduction The addition of fructose in drinking water (10% w/v) for two weeks to rats induces hypertriglyceridemia and fatty liver by reducing the expression and transcriptional activity of (..) (AU)


Subject(s)
Humans , PPAR alpha/analysis , Protein Phosphatase 2/analysis , Fructose/metabolism , Peroxisome Proliferator-Activated Receptors/analysis , Dietary Supplements/analysis , Obesity/physiopathology , Metabolic Syndrome/physiopathology
14.
J Endod ; 37(12): 1647-52, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22099898

ABSTRACT

INTRODUCTION: Tissue engineering and regenerative medicine using stem cell biology has been a promising field for treatment of local and systemic intractable diseases. Recently, stem cells from human exfoliated deciduous teeth (SHED) have been identified as a novel population of stem cells. This study focused on the characterization of SHED as compared with bone marrow-derived mesenchymal stem cells (BMMSCs). METHODS: We investigated potential characteristics of SHED by using DNA microarray, real-time reverse transcriptase polymerase chain reaction, and immunofluorescence analysis. RESULTS: Multiple gene expression profiles indicated that the expression of 2753 genes in SHED had changed by ≥2.0-fold as compared with that in BMMSCs. One of the most significant pathways that accelerated in SHED was that of bone morphogenetic protein (BMP) receptor signaling, which contains several cascades such as PKA, JNK, and ASK1. When the BMP signaling pathway was stimulated by BMP-2, the expression of BMP-2, BMP-4, Runx2, and DSPP was up-regulated significantly in SHED than that in BMMSCs. Furthermore, the BMP-4 protein was expressed much higher in SHED but not in BMMSCs, as confirmed by immunofluorescence. CONCLUSIONS: By using the gene expression profiles, this study indicates that SHED is involved in the BMP signaling pathway and suggests that BMP-4 might play a crucial role in this. These results might be useful for effective cell-based tissue regeneration, including that of bone, pulp, and dentin, by applying the characteristics of SHED.


Subject(s)
Bone Marrow Cells/physiology , Mesenchymal Stem Cells/physiology , Stem Cells/physiology , Tooth, Deciduous/cytology , Bone Morphogenetic Protein 2/analysis , Bone Morphogenetic Protein 4/analysis , Bone Morphogenetic Protein Receptors/analysis , Calcification, Physiologic/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 4/analysis , Cell Lineage , Core Binding Factor Alpha 1 Subunit/analysis , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/analysis , Extracellular Matrix Proteins/analysis , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , MADS Domain Proteins/analysis , MAP Kinase Kinase 4/analysis , MAP Kinase Kinase 6/analysis , MAP Kinase Kinase Kinase 5/analysis , MAP Kinase Signaling System/physiology , MEF2 Transcription Factors , Myogenic Regulatory Factors/analysis , Oligonucleotide Array Sequence Analysis , Peroxisome Proliferator-Activated Receptors/analysis , Phosphoproteins/analysis , Protein Kinases/analysis , Proto-Oncogene Protein c-ets-2/analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sialoglycoproteins/analysis , Signal Transduction/physiology , ets-Domain Protein Elk-4/analysis
15.
J Nutr ; 140(10): 1808-14, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20702748

ABSTRACT

Excessive and chronic alcohol intake leads to a lower hepatic vitamin A status by interfering with vitamin A metabolism. Dietary provitamin A carotenoids can be converted into vitamin A mainly by carotenoid 15,15'-monooxygenase 1 (CMO1) and, to a lesser degree, carotenoid 9'10'-monooxygenase 2 (CMO2). CMO1 has been shown to be regulated by several transcription factors, such as the PPAR, retinoid X receptor, and thyroid receptor (TR). The regulation of CMO2 has yet to be identified. The impact of chronic alcohol intake on hepatic expressions of CMO1 and CMO2 and their related transcription factors are unknown. In this study, Fischer 344 rats were pair-fed either a liquid ethanol Lieber-DeCarli diet (n = 10) or a control diet (n = 10) for 11 wk. Hepatic retinoid concentration and expressions of CMO1, CMO2, PPARγ, PPARα, and TRß as well as plasma thyroid hormones levels were analyzed. We observed that administering alcohol decreased hepatic retinoid levels but increased mRNA concentrations of CMO1, CMO2, PPARγ, PPARα, and TRß and upregulated protein levels of CMO2, PPARγ, and PPARα. There was a positive correlation of PPARγ with CMO1 (r = 0.89; P < 0.0001) and both PPARγ and PPARα with CMO2 (r = 0.72, P < 0.001 and r = 0.62, P < 0.01, respectively). Plasma thyroid hormone concentrations did not differ between the control rats and alcohol-fed rats. This study suggests that chronic alcohol intake significantly upregulates hepatic expression of CMO1 and, to a much lesser extent, CMO2. This process may be due to alcohol-induced PPARγ expression and lower vitamin A status in the liver.


Subject(s)
Ethanol/administration & dosage , Liver/enzymology , Peroxisome Proliferator-Activated Receptors/genetics , Up-Regulation/drug effects , beta-Carotene 15,15'-Monooxygenase/genetics , Animals , Fatty Acid Desaturases/genetics , Liver/chemistry , Male , PPAR alpha/analysis , PPAR alpha/genetics , PPAR gamma/analysis , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptors/analysis , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Retinoids/analysis , Thyroid Hormone Receptors beta/analysis , Thyroid Hormone Receptors beta/genetics , Thyroid Hormones/blood
16.
J. physiol. biochem ; 66(1): 29-37, mar. 2010.
Article in English | IBECS | ID: ibc-122847

ABSTRACT

No disponible


In order to evaluate the expression of nuclear receptors at the peripheral level in obese subjects,messenger RNA (mRNA) levels of different isoforms of retinoic acid receptor (RAR), triiodothyronine(TR), and peroxisome proliferator-activated receptor (PPAR) were determined and compared in peripheral mononuclear blood cells (PBMC) and subcutaneous white adipose tissue (SWAT). Twelve lean subjects and 68 obese subjects divided into weight gain (WG), weight-stable (WS), and weight loss (WL) groups were studied. Nuclear receptor mRNA levels were assessed in PBMC and SWAT using a quantitative real-time reverse transcription polymerase chain (..) (AU)


Subject(s)
Humans , Leukocytes, Mononuclear , Obesity/genetics , RNA, Messenger/analysis , Receptors, Retinoic Acid/analysis , Triiodothyronine/analysis , Peroxisome Proliferator-Activated Receptors/analysis , Subcutaneous Fat , Thyroid Hormones
17.
Rev. esp. cardiol. Supl. (Ed. impresa) ; 8(supl.E): 22e-29e, 2008. graf, tab, ilus
Article in Spanish | IBECS | ID: ibc-166422

ABSTRACT

El objetivo del tratamiento de los pacientes hipertensos no sólo es reducir las cifras de presión arterial, sino también disminuir su riesgo cardiovascular total. Es importante, por lo tanto, que los fármacos utilizados, además de su acción hipotensora, posean un perfil metabólico beneficioso. Los antagonistas de los receptores de angiotensina II (AR-II) son de demostrada eficacia antihipertensiva y también tienen efectos beneficiosos en cuanto al metabolismo de los lípidos y los hidratos de carbono y en la prevención de nuevos casos de diabetes mellitus. Estas acciones se deben al bloqueo de los receptores AT1, pero en algunos ARA-II, aunque no parece que se trate de un efecto de clase, también a través de una acción parcialmente agonista de los receptores nucleares PPARγ. Si estos efectos metabólicos beneficiosos, demostrados hasta ahora experimentalmente y en algunos estudios, se corroboran en ensayos clínicos más amplios, los convertirían en los fármacos ideales para el tratamiento de los pacientes hipertensos con otros factores de riesgo cardiovascular y/o con síndrome metabólico (AU)


The aim of treatment in hypertensive patients is not only to reduce blood pressure but also to minimize the overall cardiovascular risk. It is important, therefore, that the drugs administered have a beneficial influence on metabolic parameters as well as an antihypertensive effect. It has been shown that angiotensin-II receptor antagonists (ARA IIs) are effective antihypertensives that also have a positive influence on lipid and carbohydrate metabolism. Moreover, they can help prevent the development of new-onset diabetes. These actions arise from the blockade of angiotensin-II type-1 (AT-1) receptors, while some ARA IIs, though this does not appear to be a class effect, are also able to act like partial agonists of the nuclear receptor peroxisome proliferatoractivated receptor-gamma (PPAR-Á). If these beneficial metabolic effects, which have been found in experimental studies and in a few clinical studies, are confirmed in larger clinical trials, ARA IIs will become the ideal drugs for treating hypertensive patients with other cardiovascular risk factors or with metabolic syndrome (AU)


Subject(s)
Humans , Angiotensin II/therapeutic use , Receptor, Angiotensin, Type 2/therapeutic use , Hypotension/drug therapy , Hypotension/prevention & control , Peroxisome Proliferator-Activated Receptors/analysis , Metabolic Syndrome/complications , Diabetes Mellitus/prevention & control , Metabolic Syndrome/drug therapy , Angiotensin II/metabolism , Lipids/therapeutic use
18.
J Endocrinol ; 189(2): 199-209, 2006 May.
Article in English | MEDLINE | ID: mdl-16648288

ABSTRACT

Peroxisome proliferator-activated receptors (PPARalpha, PPARbeta/delta and PPARgamma) are a family of nuclear receptors that are activated by binding of natural ligands, such as polyunsaturated fatty acids or by synthetic ligands. Synthetic molecules of the glitazone family, which bind to PPARgamma, are currently used to treat type II diabetes and also to attenuate the secondary clinical symptoms frequently associated with insulin resistance, including polycystic ovary syndrome (PCOS). PPARs are expressed in different compartments of the reproductive system (hypothalamus, pituitary, ovary, uterus and testis). Conservative functions of PPARs in mammalian species could be suggested through several in vivo and in vitro studies, especially in the ovary and during placental development. Several groups have described a strong expression of PPARgamma in ovarian granulosa cells, and glitazones modulate granulosa cell proliferation and steroidogenesis in vitro. All these recent data raise new questions about the biologic actions of PPARs in reproduction and their use in therapeutic treatments of fertility troubles such as PCOS or endometriosis. In this review, we first describe the roles of PPARs in different compartments of the reproductive axis (from male and female gametogenesis to parturition), with a focus on PPARgamma. Secondly, we discuss the possible molecular mechanisms underlying the effect of glitazones on PCOS. Like other 'insulin sensitizer' molecules, such as metformin, glitazones may in fact act directly on ovarian cells. Finally, we discuss the eventual actions of PPARs as mediators of environmental toxic substances for reproductive function.


Subject(s)
Gametogenesis/physiology , Parturition/physiology , Peroxisome Proliferator-Activated Receptors/physiology , Corpus Luteum/metabolism , Embryonic Development/physiology , Estradiol/biosynthesis , Female , Granulosa Cells/metabolism , Humans , Hypothalamo-Hypophyseal System/chemistry , Hypothalamo-Hypophyseal System/physiology , Infertility, Female/physiopathology , Male , Ovary/chemistry , Ovary/physiology , Peroxisome Proliferator-Activated Receptors/analysis , Placenta/physiopathology , Polycystic Ovary Syndrome/physiopathology , Progesterone/biosynthesis , Testis/chemistry , Testis/physiology
19.
Reproduction ; 131(5): 917-27, 2006 May.
Article in English | MEDLINE | ID: mdl-16672356

ABSTRACT

This study documents the expression of prostacyclin (PGI2) synthase (PTGIS) and PGI2 receptors in the trophoblast and uterus of the ewe at the time of maternal recognition of pregnancy (i.e. days 7, 9, 12, 14 and 17). The membrane receptor for PGI2 (PTGIR) and the nuclear receptors, i.e. peroxisome proliferator-activated receptors (PPAR) and their heterodimer partners the retinoid X receptors (RXR), were analysed. In the endometrium, PTGIS transcript and protein were expressed at day 9 of pregnancy and levels declined from days 12 to 17. Immunohistochemistry and in situ hybridization indicated that PTGIS was mainly located in the luminal epithelium of the endometrium. Endometrial PTGIR, PPARA, PPARG and RXRG expression was regulated during the peri-implantation period whereas PPARD, RXRA and RXRB were consistently expressed. In the trophoblast, PTGIS transcript levels rose as development progressed and peaked at day 17. PTGIR and PPARA transcripts peaked before day 12 and then declined and became nearly undetectable by day 17, whereas PPARD and PPARG transcript levels rose steadily from days 12 to 17. Because the PPARs and the RXRs display different expression profiles, we suggest that different heterodimers may form and support distinct functions as development proceeds. Our results also underline the importance of PTGIS and PPARD in the trophoblast and PTGIR in the uterus, suggesting that PGI2 is of both uterine and trophoblastic origin and is involved in a complex signalling pathway at around the time of implantation in the ewe.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Embryo Implantation/physiology , Endometrium/chemistry , Gene Expression Regulation, Developmental , Intramolecular Oxidoreductases/genetics , Receptors, Epoprostenol/genetics , Trophoblasts/chemistry , Animals , Base Sequence , Blotting, Western/methods , Cattle , Cytochrome P-450 Enzyme System/analysis , DNA Probes/genetics , Female , Humans , Immunohistochemistry/methods , In Situ Hybridization/methods , Intramolecular Oxidoreductases/analysis , Molecular Sequence Data , Peroxisome Proliferator-Activated Receptors/analysis , Peroxisome Proliferator-Activated Receptors/genetics , Pregnancy , Receptors, Epoprostenol/analysis , Retinoid X Receptors/analysis , Retinoid X Receptors/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sheep
20.
Exp Gerontol ; 41(4): 372-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16600556

ABSTRACT

As ageing changes the activity of several transcription factors in the rat cortex, we were interested in determining whether similar changes also appear in the hippocampus of old rats. We determined by electrophoretic gel shift assays the binding activity of nuclear factor kappa B (NFkappaB), activator protein-1 (AP-1), peroxisome proliferator-activated receptor (PPAR), and liver X receptor (LXR) in cortex and hippocampus samples from young (3-month-old), and old (18-month-old) male and female Sprague-Dawley rats. NFkappaB activity increased in old male and female rats, though only in cortex samples, while AP-1 activity decreased only in the cortex and hippocampus of old female animals. LXR activity decreased in all conditions, except in old male cortexes; whereas PPAR activity only decreased in the hippocampus of old female rats. Decreases in AP-1 and PPAR activities restricted to old female rats did not result from an age-related decline in plasma 17beta-estradiol concentration, as their activities did not change in samples obtained from ovariectomized young female rats. Our results indicate that ageing induces a complex pattern of changes in the brain-binding activity of NFkappaB, AP-1, PPAR and LXR, depending on the anatomical origin of the samples (cortex or hippocampus), and the sex of the animals studied.


Subject(s)
Aging/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Transcription Factors/metabolism , Aging/physiology , Animals , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay/methods , Female , Liver X Receptors , Male , NF-kappa B/analysis , NF-kappa B/metabolism , Orphan Nuclear Receptors , Ovariectomy , Ovary/physiology , Peroxisome Proliferator-Activated Receptors/analysis , Peroxisome Proliferator-Activated Receptors/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factor AP-1/analysis , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...