Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 48
Filtrer
1.
Gene ; 686: 194-202, 2019 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-30340050

RÉSUMÉ

Previous studies have proposed that caloric restriction (CR) regulates many cell functions and prolongs the lifespan of an organism. Our previous studies proposed that CR also prevents follicular activation and preserves the ovarian reserve in mice by activating SIRT1. To test if SIRT1 preserves the ovarian reserve and prolongs the ovarian longevity, we generated SIRT1 knock-in mice that can overexpress SIRT1 in oocytes of the mouse. Ovaries of the mice at ages 35 days and 15 months were collected, and the follicular development and follicular reserve were examined. The vaginal opening and onset of estrus of transgenic female mice (both the homozygous and heterozygous for SIRT1 overexpression) were later than that of wild-type mice. Both the homozygous and heterozygous SIRT1-overexpressing mice had a larger and stronger reproductive capacity than wild-type mice. Moreover, 35-day-old and 15-month-old homozygous and heterozygous SIRT1-overexpressing mice also had a higher mean number and percentage of healthy follicles, fewer atretic follicles than wild-type mice, and the mean number and percentage of primordial follicles in both the homozygous and heterozygous SIRT1-overexpressing mice were higher than wild-type mice at the same age. However, the phenotypes of heterozygous and homozygous transgenic mice came no difference. Immunohistochemistry showed increased expression of SIRT1 and FOXO3a, and decreased expression of mTOR in both the homozygous and heterozygous SIRT1-overexpressing mice compared with wild-type mice. Thus, oocyte-specific SIRT1-overexpressing mice continuously activate FOXO3a and suppress mTOR and have a larger reproductive capacity, larger follicle reserve and longer ovarian lifespan.


Sujet(s)
Restriction calorique , Régulation de l'expression des gènes codant pour des enzymes , Réserve ovarienne , Ovaire/enzymologie , Sirtuine-1 , Animaux , Femelle , Protéine O3 à motif en tête de fourche/génétique , Protéine O3 à motif en tête de fourche/métabolisme , Techniques de knock-in de gènes , Souris , Souris transgéniques , Ovaire/cytologie , Sirtuine-1/biosynthèse , Sirtuine-1/génétique , Sérine-thréonine kinases TOR/biosynthèse , Sérine-thréonine kinases TOR/génétique
2.
Sci Rep ; 7(1): 14707, 2017 10 31.
Article de Anglais | MEDLINE | ID: mdl-29089617

RÉSUMÉ

The aim of this study was to determine whether low dose doxycycline as an anti-inflammatory agent could improve glucose metabolism in diabetic animals. Therefore, doxycycline was supplemented in drinking water to 6-week-old male db/db mice for 10 weeks. Doxycycline reduced perirenal/epididymal fat, Lee's index, and liver cholesterol. Blood HDL-cholesterol increased, but total cholesterol and aspartate transaminase decreased. Glucose and insulin tolerances were improved, accompanying with reduced fasting blood glucose, insulin, HOMA-IR and advanced glycation end products. Islet number, ß-cell percentage and mass increased, while islet size decreased. Consistently, less apoptosis but more ß-cell proliferation were found in islets of treated mice. Freshly isolated islets from treated mice showed higher insulin content and enhanced glucose stimulated insulin secretion (GSIS). In addition, purified islets of Balb/c mice showed increased GSIS after cultivation in vitro with doxycycline, but not with chloramphenicol and levofloxacin. Inflammation markers, including lipopolysaccharides (LPS) and C-reactive protein (CRP) in serum as well as CD68-positive cells in treated islets, decreased significantly. Finally, LPS stimulated the production of inflammatory factors but inhibited GSIS of MIN6 cells; however, the effects were completely reversed by doxycycline. The results support further study of possible long-term usage of sub-antimicrobial doxycycline in diabetic patients.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Diabète expérimental/traitement médicamenteux , Diabète de type 2/traitement médicamenteux , Doxycycline/usage thérapeutique , Glucose/métabolisme , Inflammation/traitement médicamenteux , Cellules à insuline/physiologie , Insuline/métabolisme , Foie/physiologie , Animaux , Apoptose , Prolifération cellulaire , Cellules cultivées , Produits terminaux de glycation avancée/métabolisme , Humains , Sécrétion d'insuline , Cellules à insuline/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Mâle , Souris , Souris de lignée BALB C , Souches mutantes de souris
3.
J Microbiol Biotechnol ; 27(6): 1098-1105, 2017 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-28297751

RÉSUMÉ

Vesicular stomatitis virus G glycoprotein (VSV-G) has been widely used for pseudotyping retroviral, lentiviral, and artificial viral vectors. The objective of this study was to establish a potential approach for large-scale production of VSV-G. To this end, VSV-G was cloned with an N-terminal His-tag into Pichia pastoris expression vector pPIC3.5K. Three clones (Muts) containing the VSV-G expression cassette were identified by PCR. All clones proliferated normally in expansion medium, whereas the proliferation was reduced significantly under induction conditions. VSV-G protein was detected in cell lysates by western blot analysis, and the highest expression level was observed at 96 h post induction. VSV-G could also be obtained from the condition medium of yeast protoplasts. Furthermore, VSV-G could be incorporated into Ad293 cells and was able to induce cell fusion, leading to the transfer of cytoplasmic protein. Finally, VSV-G-mediated DNA transfection was assayed by flow cytometry and luciferase measurement. Incubation of VSV-G lysate with the pGL3-control DNA complex increased the luciferase activity in Ad293 and HeLa cells by about 3-fold. Likewise, incubation of VSV-G lysate with the pCMV-DsRed DNA complex improved the transfection efficiency into Ad293 by 10% and into HeLa cells by about 1-fold. In conclusion, these results demonstrate that VSV-G could be produced from P. pastoris with biofunctionalities, demonstrating that large-scale production of the viral glycoprotein is feasible.


Sujet(s)
ADN , Glycoprotéines membranaires/biosynthèse , Glycoprotéines membranaires/génétique , Pichia/génétique , Transfection , Protéines de l'enveloppe virale/biosynthèse , Protéines de l'enveloppe virale/génétique , Animaux , Numération cellulaire , Expression des gènes , Vecteurs génétiques , Cellules HeLa , Humains , Pichia/composition chimique , Pichia/croissance et développement , Protoplastes , Protéines recombinantes/biosynthèse , Transfection/méthodes
4.
Appl Biochem Biotechnol ; 181(2): 748-761, 2017 Feb.
Article de Anglais | MEDLINE | ID: mdl-27665615

RÉSUMÉ

The objective of this study was to formulate a novel gene delivery system based on the erythrocyte ghost (EG) integrated with fusogenic viral glycoprotein vesicular stomatitis virus glycoprotein G (VSV-G). VSV-G proteins were harvested as condition medium of Ad293 cells carrying a VSV-G transgene and then incorporated into EG. Plasmid DNA was condensed by various transfection reagents. A luciferase expression construct (pGL3-control) and a DsRed expression cassette (pCMV-DsRed) were used to evaluate the delivery efficiency of DNA/EG/VSV-G complexes. VSV-G proteins could be incorporated into EG in static incubation under acidic conditions as evidenced by the Western blot analysis. Condensed plasmid DNA was bound mostly to the outer surface of EG, which could be detected by electromicroscopy and measured by electrophoresis. EG/VSV-G complexes stimulated the delivery of pGL3-control into Ad293 cells significantly with the luciferase activity increased about 4-fold as compared to that of the control. The delivery of pCMV-DsRed was also enhanced with the percentage of DsRed-positive Ad293 cells increased from 55 % to about 80 %. Moreover, the transfection efficiency in 3T3, HeLa, INS-1, and bone marrow stem cell (BMSC) cells increased about 2-3-fold. Finally, confocal microscopy analysis showed that incorporation of VSV-G significantly enhanced the endocytosis of EG into target cells. In the present study, a novel type of non-viral DNA delivery vehicle consisting of EG and fusogenic VSV-G proteins was formulated, which showed superior transfection efficiency even in cells resistant to classical transfection.


Sujet(s)
ADN/génétique , Membrane érythrocytaire/génétique , Amélioration génétique/méthodes , Glycoprotéines/génétique , Lentivirus/génétique , Transfection/méthodes , Cellules 3T3 , Animaux , ADN/administration et posologie , Cellules HeLa , Humains , Souris
5.
Gen Comp Endocrinol ; 237: 98-108, 2016 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-27570060

RÉSUMÉ

OBJECTIVES: We previously found niacin receptor GPR109A was expressed in murine islet beta-cells, and signaling through GPR109A inhibited glucose stimulated insulin secretion (GSIS). However, the expression of GPR109A in human islets and its functional relevance is still not known. METHODS: The expression of GPR109A was examined by antibody staining and in situ hybridization on pancreatic paraffin sections. GPR109A was cloned and expressed in INS-1 islet beta-cells. Intracellular cAMP and GSIS were determined using enzyme-linked immunosorbent assay (ELISA). RESULTS: The expression of GPR109A was confirmed in murine islet beta-cells and further detected in human counterparts by using commercially available polyclonal antibodies. In situ hybridization study detected the transcripts of GPR109A, but not that of closely related GPR109B. Furthermore, GPR109A was significantly reduced in islets from diabetic individuals and animal model of db/db mice as compared to their respective controls. Further, GPR109A levels in insulinoma were also reduced dramatically as compared to islets found in corresponding non-tumor normal tissues. Quantitative RT-PCR analysis demonstrated that GPR109A transcripts were severely down-regulated in rodent insulinoma cell lines as compared to that of freshly isolated islets from mice. Finally, human and murine GPR109A expression cassettes were transfected into INS-1 cells, which resulted in reduced accumulation of cAMP and insulin secretion after incubation with niacin. The effect could be completely abrogated by pretreatment with pertussis toxin. CONCLUSIONS: These results demonstrate that GPR109A is functionally expressed in both human and murine islet beta-cells. However, the role of GPR109A in the prevention of diabetes or insulinoma needs further study.


Sujet(s)
Diabète de type 2/métabolisme , Régulation négative , Cellules à insuline/métabolisme , Insuline/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Récepteurs nicotiniques/métabolisme , Sujet âgé , Animaux , AMP cyclique/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Femelle , Technique d'immunofluorescence , Glucose/pharmacologie , Humains , Sécrétion d'insuline , Cellules à insuline/effets des médicaments et des substances chimiques , Insulinome/métabolisme , Mâle , Souris , Souris de lignée BALB C , Adulte d'âge moyen , ARN messager/génétique , ARN messager/métabolisme , Récepteurs couplés aux protéines G/génétique , Récepteurs nicotiniques/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique
6.
Biomed Microdevices ; 18(3): 41, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-27165101

RÉSUMÉ

The objective of this study was to determine if plasma membrane vesicles (PMVs) could be exploited for efficient transfer of macro-biomolecules and mitochondria. PMVs were derived from mechanical extrusion, and made fusogenic (fPMVs) by incorporating the glycoprotein G of vesicular stomatitis virus (VSV-G). Confocal microscopy examination revealed that cytoplasmic proteins and mitochondria were enclosed in PMVs as evidenced by tracing with cytoplasmically localized and mitochondria-targeted EGFP, respectively. However, no fluorescence signal was detected in PMVs from cells whose nucleus was labeled with an EGFP-tagged histone H2B. Consistently, qRT-PCR measurement showed that mRNA, miRNA and mitochondrial DNA decreased slightly; while nuclear DNA was not measureable. Further, Western blot analysis revealed that cytoplasmic and membrane-bound proteins fell inconspicuously while nuclear proteins were barely detecsle. In addition, fPMVs carrying cytoplamic DsRed proteins transduced about ~40 % of recipient cells. The transfer of protein was further confirmed by using the inducible Cre/loxP system. Mitochondria transfer was found in about 20 % recipient cells after incubation with fPMVs for 5 h. To verify the functionalities of transferred mitochondria, mitochodria-deficient HeLa cells (Rho0) were generated and cultivated with fPMVs. Cell enumeration demonstrated that adding fPMVs into culture media stimulated Rho0 cell growth by 100 % as compared to the control. Lastly, MitoTracker and JC-1 staining showed that transferred mitochondria maintained normal shape and membrane potential in Rho0 cells. This study established a time-saving and efficient approach to delivering proteins and mitochondria by using fPMVs, which would be helpful for finding a cure to mitochondria-associated diseases. Graphical abstract Schematic of the delivery of macro-biomolecules and organelles by fPMVs. VSV-G-expressing cells were extruded through a 3 µm polycarbonate membrane filter to generate fusogenic plasma membrane vesicles (fPMVs), which contain bioactive molecules and organelles but not the nucleus. fPMVs can be endocytosed by target cells, while the cargo is released due to low-pH induced membrane fusion. These nucleus-free fPMVs are efficient at delivery of cytoplasmic proteins and mitochondria, leading to recovery of mitochondrial biogenesis and proliferative ability in mitochondria-deficient cells.


Sujet(s)
Membrane cellulaire/métabolisme , Glycoprotéines membranaires/métabolisme , Mitochondries/métabolisme , Vésicules de transport/métabolisme , Protéines de l'enveloppe virale/métabolisme , Lignée cellulaire , Noyau de la cellule , ADN mitochondrial/génétique , Génomique , Protéines à fluorescence verte/métabolisme , Cellules HeLa , Histone/métabolisme , Humains , microARN/génétique , Ciment carboxylate/composition chimique , ARN messager/génétique , Analyse de séquence d'ADN , Virus de la stomatite vésiculeuse de type Indiana
7.
PLoS One ; 10(6): e0129092, 2015.
Article de Anglais | MEDLINE | ID: mdl-26046525

RÉSUMÉ

Ectopically expressed Cre recombinase in extrapancreatic tissues in RIP-Cre mice has been well documented. The objective of this study was to find a simple solution that allows for improved beta-cell specific targeting. To this end, the RIP-Cre and reporter CMV-loxP-DsRed-loxP-EGFP expression cassettes were configurated into a one-plasmid and two-plasmid systems, which labeled approximately 80% insulin-positive INS-1 cells after 48 h transfection. However, off-target labeling was robustly found in more than 15% insulin-negative Ad293 cells. When an IRES element was inserted in front of Cre to reduce the translation efficiency, the ratio of recombination between INS-1 and Ad293 cells increased 3-4-fold. Further, a series of Cre mutants were generated by site-directed mutagenesis. When one of the mutants, Cre(H289P) in both configurations, was used in the experiment, the percentage of recombination dropped to background levels in a number of insulin-negative cell lines, but decreased only slightly in INS-1 cells. Consistently, DNA substrate digestion assay showed that the enzymatic activity of Cre(H289P) was reduced by 30-fold as compared to that of wild-type. In this study, we reported the generation of constructs containing RIP and Cre mutants, which enabled enhanced beta-cell specific labeling in vitro. These tools could be invaluable for beta-cell targeting and to the study of islet development.


Sujet(s)
Cellules à insuline/cytologie , Insuline/génétique , Integrases/génétique , Animaux , Lignée cellulaire , Technique d'immunofluorescence , Gènes rapporteurs , Protéines à fluorescence verte/analyse , Protéines à fluorescence verte/génétique , Humains , Cellules à insuline/métabolisme , Integrases/métabolisme , Mutagenèse dirigée , Plasmides/génétique , Mutation ponctuelle , Régions promotrices (génétique) , Rats , Coloration et marquage , Transfection
8.
Eur J Med Res ; 20: 22, 2015 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-25889584

RÉSUMÉ

BACKGROUND: Silent information regulator 2 related enzyme 1 (SIRT1) is one of the key factors in the mechanism of calorie restriction (CR) extending lifespan of animals. The aim of the study is to investigate if CR prolongs ovarian lifespan in mice through activating SIRT1 signaling. METHODS: In the present study, 21 female C57BL/6 mice were divided into three groups: the control (n = 7), CR (n = 7), and SRT1720 (n = 7) groups. After the 26-week treatment, the number of ovarian follicles at each stage was counted, and Western blot was performed. RESULTS: The number of surviving follicles in ovaries of the SRT1720 group was less than that of the CR group but more than that of the normal control (NC) group. The number of atretic follicles in the ovaries of the SRT1720 group was similar to that of the CR group but less than that of the NC group. The number of primordial follicles in the ovaries of the SRT1720 group was less than that of the CR group but more than that of the NC group. The numbers of primary follicles, secondary follicles, antral follicles, and corpora lutea in the SRT1720 group were similar to those in the CR group. Western blot analysis showed that the expression of SIRT1, SIRT6, FOXO3a, and NRF1 proteins was upregulated, and p53 was downregulated in both the CR group and the SRT1720 group compared to the control group. CONCLUSIONS: Our results indicate that CR inhibits the activation of primordial follicles and development of follicles at different stages, thus preserving the reserve of follicle pool (at least partly) through activating SIRT1 signaling.


Sujet(s)
Restriction calorique , Follicule ovarique/métabolisme , Transduction du signal , Sirtuine-1/métabolisme , Animaux , Femelle , Protéine O3 à motif en tête de fourche , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme , Souris , Souris de lignée C57BL , Facteur nucléaire-1 respiratoire/génétique , Facteur nucléaire-1 respiratoire/métabolisme , Follicule ovarique/croissance et développement , Sirtuine-1/génétique , Sirtuines/génétique , Sirtuines/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
9.
Reprod Sci ; 22(1): 60-7, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25001019

RÉSUMÉ

Caloric restriction (CR) is known to increase the number of primordial follicles and prolong the reproductive life span. However, how CR modulates follicular development is not well understood. In the present study, we examined the effects of CR on follicular development in rats and investigated the underlying mechanism. After 10 weeks of CR or high-fat diet, ovarian follicles at different developmental stages were examined by histological analysis. Plasma levels of luteinizing hormone (LH), follicle-stimulating hormone (FSH), and estrogen (ESG) were measured, and the levels of mammalian target of rapamycin (mTOR), p70S6 kinase (p70S6K), and phosphorylated p70S6K in the ovary were detected by Western blot. The results showed that the reserve of follicle pool in CR rats was increased, accompanied by decreased level of phosphorylated p70S6K in the ovary, and decreased serum LH, FSH, and ESG levels. Taken together, these results suggest that CR may suppress ovarian follicular development and enhance the follicle pool reserve by inhibiting mTOR signaling.


Sujet(s)
Restriction calorique , Prolifération cellulaire , Follicule ovarique/enzymologie , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme , Phénomènes physiologiques nutritionnels chez l'animal , Animaux , Poids , Alimentation riche en graisse , Oestrogènes/sang , Femelle , Hormone folliculostimulante/sang , Hormone lutéinisante/sang , État nutritionnel , Phosphorylation , Rat Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/métabolisme , Facteurs temps
10.
Ann Clin Lab Sci ; 44(4): 410-8, 2014.
Article de Anglais | MEDLINE | ID: mdl-25361925

RÉSUMÉ

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) has become the most common form of chronic liver disease in the world. Recent studies in cultured cells and mice have shown that sirtuin, especially sirtuin 1 (SIRT1), is a key metabolic sensor for regulating metabolic homeostasis and thus has the potential to ameliorate NAFLD. For the purposes of this study, we hypothesized that the inhibition of sirtuin signaling might contribute to the development of NAFLD. METHODS: Tissue was obtained from hepatectomy specimens (10 samples), and medicolegal autopsies (10 samples). Liver tissue sections were stained with H&E. Expression of sirtuin in liver tissues in NAFLD and control group was investigated by RT-PCR and Western blotting. RESULTS: RT-PCR and Western blotting demonstrated decreased expression of SIRT1, SIRT3, SIRT5, and SIRT6 in the NAFLD group in comparison with the control group. Increased expression of lipogenic genes including sterol regulatory element binding protein-1 (SREBP-1), fatty acid synthase (FASN), and acetyl-CoA carboxylase (ACC) was noted within the NAFLD group. In contrast to the other SIRT genes, the expression of SIRT4 was upregulated. CONCLUSION: Our study provides direct evidence of the downregulation of sirtuin signaling that suppresses lipid synthesis in the liver of NAFLD patients, which may promote NAFLD development.


Sujet(s)
Régulation négative/physiologie , Foie/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/physiopathologie , Sirtuines/métabolisme , Acetyl-coA carboxylase/génétique , Acetyl-coA carboxylase/métabolisme , Fatty acid synthase type I/génétique , Fatty acid synthase type I/métabolisme , Femelle , Humains , Foie/anatomopathologie , Mâle , ARN messager/métabolisme , Sirtuines/génétique , Protéine-1 de liaison à l'élément de régulation des stérols/génétique , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme
11.
J Ovarian Res ; 7: 97, 2014 Oct 21.
Article de Anglais | MEDLINE | ID: mdl-25330910

RÉSUMÉ

BACKGROUND: The prevalence of obesity is increasing worldwide and significantly affects fertility and reproduction in both men and women. Our recent study has shown that excess body fat accelerates ovarian follicle development and follicle loss in rats. The aim of the present study is to explore the effect of SIRT1 activator SRT1720 on the reserve of ovarian follicle pool and ovarian lifespan of obese mice and the underlying mechanism associated with SIRT1 and mTOR signaling. METHODS: Adult female Kunming mice (n = 36) were randomly divided into three groups: the normal control (NC) group (n = 8), the caloric restriction (CR) group (fed 70% food of the NC group, n = 8) and the high-fat diet (HF) group (fed a rodent chow containing 20% fat, n = 20). After 4 months, the HF mice were further randomly divided into three groups: the control high-fat diet (CHF, n = 8) group (treated every day with an intraperitoneal injection of vehicle), the SRT1720 (SRT, n = 6) group (treated every other day with an intraperitoneal injection of SRT1720 (50 mg/kg)), the SRT1720 and nicotinamide (NAM, n = 6) group (treated every other day with an intraperitoneal injection of SRT1720 (50 mg/kg) and every day with an intraperitoneal injection of nicotinamide (100 mg/kg)). After 6 weeks of treatment, ovaries were harvested for histological and Western blotting analyses. RESULTS: The body weight, ovary weight and visceral fat in the SRT group were significantly lower than those in the CHF group at the end of treatment. Histological analysis showed that the SRT mice had significantly greater number and percentage of primordial follicles, but lower number and percentage of corpora lutea and atretic follicles than the CHF mice and NAM mice. Western blot analysis demonstrated that the levels of SIRT1, SIRT6, FOXO3a and NRF-1 protein expression significantly increased in the ovaries of SRT mice, whereas those of mTORC1, p-mTOR, p-p70S6K, NFκB and p53 decreased compared to the CHF and NAM mice. CONCLUSIONS: Our study suggests that SRT1720 may improve the follicle pool reserve in HF diet-induced obese female mice via activating SIRT1 signaling and suppressing mTOR signaling, thus extending the ovarian lifespan.


Sujet(s)
Fécondostimulants féminins/pharmacologie , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Infertilité féminine/traitement médicamenteux , Obésité/complications , Sirtuine-1/métabolisme , Animaux , Alimentation riche en graisse/effets indésirables , Activateurs d'enzymes/pharmacologie , Cycle oestral/effets des médicaments et des substances chimiques , Femelle , Infertilité féminine/étiologie , Graisse intra-abdominale/anatomopathologie , Souris , Taille d'organe , Réserve ovarienne/effets des médicaments et des substances chimiques , Ovaire/effets des médicaments et des substances chimiques , Ovaire/enzymologie , Ovaire/anatomopathologie , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme
12.
Mol Med Rep ; 9(4): 1218-24, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24535021

RÉSUMÉ

Calorie restriction (CR) has been shown to increase longevity and mitigate age­associated diseases in various organisms. Numerous studies have identified that sirtuin 1 (SIRT1) is upregulated by CR. However, the expression of SIRT isoforms 2­7 in response to CR in cardiomyocytes has yet to be elucidated. Therefore, the present study aimed to investigate the cellular localization and expression of SIRT1­7 in cardiomyocytes. Twenty SD rats were fed either ad libitum (AL) or a CR diet (60% of AL) for three weeks. In addition, H9c2 cells were cultured in Dulbecco's modified Eagle medium (DMEM) supplemented with either normal­ (4.5 g/l) or low­ (1 g/l) glucose concentrations for 24 h, representing control or CR cells, respectively. CR rats were observed to have significantly lower heart and body weights (BWs) than control rats. Moreover, immunohistochemical analyzes revealed that SIRT1, 3­5 and 7 demonstrated similar localization patterns in H9c2 cells and rat cardiac tissues, with SIRT1 and 7 predominantly located in the nucleus and SIRT3­5 in the cytoplasm. This was in contrast with SIRT2, which was detected exclusively in the cytoplasm in rat cardiac tissues, but was found predominantly in the nucleus in H9c2 cells. The converse was observed for SIRT6. Quantitative polymerase chain reaction revealed that the mRNA expression of SIRT1­4 and ­7 was increased in the CR group. Western blot analysis further revealed that the protein expression of SIRT1­4 and ­7 was significantly increased in the cardiac tissues of rats in the CR group. These results suggest that CR may attenuate age­associated changes through reducing BW. Moreover, short­term CR may activate SIRT1 as well as SIRT2­4 and ­7 expression in cardiomyocytes in vivo and in vitro.


Sujet(s)
Restriction calorique , Myocytes cardiaques/métabolisme , Sirtuines/métabolisme , Animaux , Technique de Western , Poids , Lignée cellulaire , Régime alimentaire , Femelle , Régulation de l'expression des gènes , Mâle , Myocarde/métabolisme , Taille d'organe , Transport des protéines , ARN messager/génétique , ARN messager/métabolisme , Rats , Rat Sprague-Dawley , Sirtuines/génétique , Facteurs temps
13.
Mol Biol Rep ; 41(4): 2143-9, 2014.
Article de Anglais | MEDLINE | ID: mdl-24442318

RÉSUMÉ

Clodronate liposome injection is an effective approach to selectively and specifically depleting macrophages. Macrophages play a crucial role in cutaneous wound healing and are associated with excessive scar formation. Use of clodronate liposomes to enhance cutaneous wound healing and reduce scar formation could represent a major advance in wound therapy and hypertrophic scar treatment. This study aimed to investigate the effects of subcutaneous or intraperitoneal injection of clodronate liposomes on cutaneous wound healing and scar formation. A burn injury mouse model was used. Mice were treated with subcutaneous or intraperitoneal injection of clodronate liposomes. Wound healing time was analyzed and scar tissues were harvested for hematoxylin and eosin (HE) staining, reverse transcription polymerase chain reaction (RT-PCR) and Western blot analyses. Wound healing time in treated mice was extended. HE showed that the basal layer of the epidermis in treated scars was flattened, the dermis layer was not significantly thickened, and collagen fibers were well arranged, with few cells and micro vessels. RT-PCR and Western blot analyses showed that the levels of TGF-ß1 and collagen I-α2 were decreased in treated mice. Clodronate liposomes reduce excessive scar formation and delay cutaneous wound healing possibly by reducing collagen deposition and macrophage-derived TGF-ß1 expression.


Sujet(s)
Brûlures/métabolisme , Brûlures/anatomopathologie , Cicatrice/métabolisme , Acide clodronique/administration et posologie , Collagène/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Animaux , Brûlures/traitement médicamenteux , Cicatrice/traitement médicamenteux , Cicatrice/anatomopathologie , Collagène de type I/métabolisme , Modèles animaux de maladie humaine , Liposomes , Macrophages/immunologie , Macrophages/anatomopathologie , Souris , Facteurs temps , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Cicatrisation de plaie/immunologie
14.
Metabolism ; 63(1): 94-103, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24135502

RÉSUMÉ

OBJECTIVE: Studies have shown that excess body fat negatively affects reproductive functions in females. However, whether obesity affects the ovarian follicle development and ovarian lifespan and the underlying mechanism has not been well elucidated. The aim of the present study was to investigate the association between obesity and ovarian follicle development. METHODS: Adult female Sprague-Dawley rats (n = 36) were randomly divided into three groups: the normal control (NC) group, the caloric restriction (CR) group (fed 70% food of the NC group) and the high-fat diet (HF) group. They were maintained on these regimens for 18 weeks. RESULTS: The body weight, ovary weight and visceral fat in the HF group were significantly higher than those in the NC group and the CR group at the end of treatment. Histological analysis showed that the HF rats had significantly less number and percentage of primordial follicles, but greater number and percentage of developing and atretic follicles than the NC rats and CR rats. Western blot analysis demonstrated that the level of mTORC1 and p-S6K1 proteins significantly increased in the ovaries of HF rats, whereas that of SIRT1, SIRT6, FOXO3a and NRF-1 decreased compared to the NC rats. In contrast, the expression of mTORC1 and p-S6K1 dramatically declined, while that of SIRT1, SIRT6, FOXO3a and NRF1 increased in the ovaries of CR rats. CONCLUSIONS: Our study suggests that the HF diet induced obesity may accelerate the ovarian follicle development and rate of follicle loss through activating mTOR and suppressing SIRT1 signaling, thus leading to POF, and that CR may inhibit the activation of primordial follicles, follicular development and loss, thus extending the ovarian lifespan through suppressing mTOR and activating SIRT1 signaling.


Sujet(s)
Obésité/métabolisme , Obésité/anatomopathologie , Follicule ovarique/anatomopathologie , Sirtuine-1/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Animaux , Technique de Western , Restriction calorique , Alimentation riche en graisse , Femelle , Immunohistochimie , Graisse intra-abdominale/métabolisme , Follicule ovarique/croissance et développement , Répartition aléatoire , Rats , Rat Sprague-Dawley , Transduction du signal
16.
Mol Med Rep ; 8(1): 23-8, 2013 Jul.
Article de Anglais | MEDLINE | ID: mdl-23652462

RÉSUMÉ

Atherosclerosis is a chronic immunoinflammatory disease associated with blood lipid disorders. Previous studies in mice have demonstrated that liver X receptor (LXR)­ATP­binding cassette (ABC) A1/ABCG1/C­C chemokine receptor type 7 (CCR7) and nuclear factor κB (NF­κB) signaling pathways are important for atherosclerotic plaque formation. In addition, Sirtuin 1 (SIRT1) has been reported as a key regulator in the protection from risk of atherosclerosis. However, the exact mechanism by which SIRT1 prevents atherosclerosis remains largely unknown. To explore the possible mechanisms, the expression of SIRT1 and the association between SIRT1, LXR and NF­κB in the process of foam cell formation was investigated in an in vitro human mononuclear U937 cell line. Monocyte­derived foam cells were induced by palmitate and Ox­LDL treatment. Oil Red O staining revealed an accumulation of a large number of lipid droplets in foam cells. Results of reverse transcription polymerase chain reaction (RT-PCR) revealed that SIRT1 expression was downregulated during foam cell formation. In addition, the expression of LXRα and its targets, ABCA1, ABCG1 and CCR7, were downregulated. However, NF­κB and its targets, tumor necrosis factor α (TNFα) and interleukin (IL)­1ß, were upregulated in foam cells. Following activation of SIRT1 by SRT1720, the expression of LXRα and its targets increased, whereas expression of NF­κB and its targets decreased. Furthermore, the formation of foam cells was blocked. The SIRT1 inhibitor, nicotinamide, was found to eliminate the effects of SRT1720. Results of the present study indicate that SIRT1 may prevent the formation and progression of atherosclerosis by enhancing the LXR­ABCA1/ABCG1/CCR7 and inhibiting the NF­κB pathways.


Sujet(s)
Athérosclérose/génétique , Athérosclérose/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Récepteurs nucléaires orphelins/métabolisme , Transduction du signal , Sirtuine-1/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules spumeuses/effets des médicaments et des substances chimiques , Cellules spumeuses/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Récepteurs hépatiques X , Facteur de transcription NF-kappa B/génétique , Récepteurs nucléaires orphelins/génétique , Palmitates/pharmacologie , ARN messager/génétique , ARN messager/métabolisme , Sirtuine-1/métabolisme , Triglycéride/métabolisme , Cellules U937
17.
Gene ; 523(1): 82-7, 2013 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-23566837

RÉSUMÉ

To maintain the normal length of female reproductive life, the majority of primordial follicles must be maintained in a quiescent state for later use. In this study, we aimed to study the effects of rapamycin on primordial follicle development and investigate the role of mTOR and sirtuin signaling. Rats were treated every other day with an intraperitoneal injection of rapamycin (5mg/kg) or vehicle. After 10weeks of treatment, ovaries were harvested for hematoxylin and eosin (HE) staining, and analysis by immunohistochemistry and Western blotting. HE staining showed that the number and percentage of primordial follicles in the rapamycin-treated group were twice the control group (P<0.001). Immunohistochemical analysis showed that mTOR and phosphorylated-p70S6K were extensively expressed in surviving follicles with strong staining observed in the cytoplasm of the oocyte. Western blotting showed decreased expression of phosphorylated mTOR and phosphorylated p70S6K in the rapamycin-treated group, and increased the expression of both SIRT1 and SIRT6 compared to the control group (P<0.05). Taken together, these results suggest that rapamycin may inhibit the transition from primordial to developing follicles and preserve the follicle pool reserve, thus extending the ovarian lifespan of female rats via the modulation of mTOR and sirtuin signalings.


Sujet(s)
Follicule ovarique/effets des médicaments et des substances chimiques , Sirolimus/pharmacologie , Sirtuine-2/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Animaux , Poids/effets des médicaments et des substances chimiques , Cytoplasme/métabolisme , Consommation alimentaire/effets des médicaments et des substances chimiques , Éosine jaunâtre/métabolisme , Cycle oestral/physiologie , Femelle , Fécondité/effets des médicaments et des substances chimiques , Hématoxyline/métabolisme , Mâle , Ovocytes/métabolisme , Taille d'organe , Follicule ovarique/métabolisme , Follicule ovarique/physiologie , Phosphorylation , Rats , Rat Sprague-Dawley , Ribosomal Protein S6 Kinases/génétique , Ribosomal Protein S6 Kinases/métabolisme , Transduction du signal , Sirtuine-2/génétique , Sirtuines/génétique , Sirtuines/métabolisme , Sérine-thréonine kinases TOR/génétique , Facteurs temps
18.
Mol Biol Rep ; 40(4): 3373-80, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23292098

RÉSUMÉ

Sirtuin 1 (SIRT1) is one member of the silent information regulator 2 (Sir2)-like family of proteins involved in glucose homeostasis in mammals. It has been reported that SIRT1 modulates endocrine signaling of glucose and fat homeostasis by regulating transcription factors such as forkhead transcription factor 3a (FOXO3a), glucose transporter 4 (GLUT4), peroxisome proliferator-activated receptor gamma (PPARγ) and PPARγ coactivator (PGC-1α). However, it is still not clear how SIRT1 is involved in the development of insulin resistance. To determine the location and expression of SIRT1 and its target proteins in rats and analyze the interactions and functions of these proteins in insulin resistance. Forty-eight male Sprague-Dawley rats were randomly divided into four regimen groups: normal control (NC), calorie restriction (CR), high-fat (HFa), and high-fructose (HFr). Animals were fed for 12 weeks and blood samples collected from tail veins at weeks 2, 4, 6, 8 and 12 after fasting for 16 h. Baseline metabolic parameters such as fasting blood sugar, insulin, cholesterol and triglycerides were analyzed. A glucose tolerance test was carried out at the end of the study. Visceral fat, consisting of epididymis and perirenal fat, was isolated and weighed. The pancreas from each animal was also immediately removed. Immunohistochemical staining was performed to detect the locations of SIRT1, FOXO3a, GLUT4, PPARγ and PGC-1α in the ß-cell of the rat pancreas. Expression in the pancreas was analyzed by western blotting. Blood biochemical analysis indicated that the HFa and HFr groups were insulin-resistant. Immunohistochemical staining showed that GLUT4 was a nuclear protein. SIRT1, FOXO3a, PPARγ and PGC-1α were present in both the nucleus and the cytoplasm of ß-cells of pancreatic islets. The expression of SIRT1, GLUT4 and PGC-1α increased significantly in response to CR, but decreased in the HFr and HFa groups. FOXO3a was similar in the CR and the NC groups, whereas it declined in the HFa and HFr groups. PPARγ was elevated in the HFa group, but dropped in the CR and HFr groups. These data suggest that SIRT1 and its regulators are involved in the development of insulin resistance.


Sujet(s)
Facteurs de transcription Forkhead/métabolisme , Transporteur de glucose de type 4/métabolisme , Insulinorésistance/génétique , Récepteur PPAR gamma/métabolisme , Protéines de liaison à l'ARN/métabolisme , Sirtuine-1/métabolisme , Facteurs de transcription/métabolisme , Animaux , Restriction calorique , Alimentation riche en graisse , Protéine O3 à motif en tête de fourche , Fructose/administration et posologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Cellules à insuline/ultrastructure , Mâle , Pancréas/cytologie , Pancréas/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Liaison aux protéines , Rats , Rat Sprague-Dawley
19.
Biochem Biophys Res Commun ; 430(2): 798-803, 2013 Jan 11.
Article de Anglais | MEDLINE | ID: mdl-23201401

RÉSUMÉ

Oxidative stress-mediated cell death in cardiomyocytes reportedly plays an important role in many cardiac pathologies. Our previous report demonstrated that mitochondrial SIRT3 plays an essential role in mediating cell survival in cardiac myocytes, and that resveratrol protects cardiomyocytes from oxidative stress-induced apoptosis by activating SIRT3. However, the exact mechanism by which SIRT3 prevents oxidative stress remains unknown. Here, we show that exposure of H9c2 cells to 50 µM H(2)O(2) for 6h caused a significant increase in cell death and the down-regulation of SIRT3. Reactive oxygen species (ROS)-mediated NF-κB activation was involved in this SIRT3 down-regulation. The SIRT3 activator, resveratrol, which is considered an important antioxidant, protected against H(2)O(2)-induced cell death, whereas the SIRT inhibitor, nicotinamide, enhanced cell death. Moreover, resveratrol negatively regulated H(2)O(2)-induced NF-κB activation, whereas nicotinamide enhanced H(2)O(2)-induced NF-κB activation. We also found that SOD2, Bcl-2 and Bax, the downstream genes of NF-κB, were involved in this pathological process. These results suggest that SIRT3 protects cardiomyocytes exposed to oxidative stress from apoptosis via a mechanism that may involve the NF-κB pathway.


Sujet(s)
Apoptose/physiologie , Myocytes cardiaques/physiologie , Facteur de transcription NF-kappa B/métabolisme , Stress oxydatif , Sirtuine-3/métabolisme , Animaux , Apoptose/génétique , Lignée cellulaire , Régulation de l'expression des gènes , Gènes bcl-2/génétique , Peroxyde d'hydrogène/pharmacologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Nicotinamide/pharmacologie , Rats , Espèces réactives de l'oxygène/métabolisme , Sirtuine-3/antagonistes et inhibiteurs , Sirtuine-3/génétique , Superoxide dismutase/génétique , Protéine Bax/génétique
20.
Aging Clin Exp Res ; 24(2): 125-33, 2012 Apr.
Article de Anglais | MEDLINE | ID: mdl-21502801

RÉSUMÉ

BACKGROUND AND AIMS: Caloric restriction (CR) extends mammals' lifespans and suppresses ovary development. Sirtuins are involved in these mechanisms. If, and to what extent CR affects ovarian lifespan and follicle development is largely unknown. We investigated the effects of moderate and severe caloric restriction compared with a high-fat dietary regimen on ovarian follicle reserves in rats. METHODS: Female Sprague-Dawley rats (n=48) randomly divided into four groups including normal control (NC), 25% caloric restriction (MCR), 45% CR (SCR) and high-fat diet (HF) were maintained on these regimens for 2 months. RESULTS: Histological analysis showed that both the 25 and 45% CR rats had a significantly higher percentage of primordial follicles and a larger number of healthy follicles than the NC rats, whereas the HF rats did not differ significantly from the NC rats. Immunohistochemical analysis revealed that SIRT1 and SIRT6 proteins were present in the nucleus and cytoplasm of the oocytes. The 25% CR diet increased the expression of both SIRT1 and SIRT6 in the ovary, whereas the 45% CR and HF diets caused a decrease in SIRT1 expression. The level of SIRT6 protein did not change with the 45% CR diet, and it appeared slightly lower in the HF than in the NC groups. CONCLUSIONS: Caloric restriction may inhibit the transition from primordial to developing follicles and extend the entire growth phase of a follicle to preserve the reserve of germ cells. SIRT1 and SIRT6 are both associated with these effects.


Sujet(s)
Restriction calorique , Alimentation riche en graisse , Follicule ovarique/métabolisme , Sirtuine-1/biosynthèse , Sirtuines/biosynthèse , Animaux , Poids/physiologie , Cholestérol/sang , Ration calorique/physiologie , Femelle , Follicule ovarique/anatomopathologie , Rats , Rat Sprague-Dawley , Sirtuine-1/métabolisme , Sirtuines/métabolisme , Triglycéride/sang
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...