Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Hum Reprod ; 31(6): 1315-26, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27052498

RESUMEN

STUDY QUESTION: What effect does diet-induced obesity have on endometrial stromal cell (ESC) decidualization? SUMMARY ANSWER: Diet-induced obesity impairs ESC decidualization. WHAT IS KNOWN ALREADY: Decidualization is important for successful implantation and subsequent health of the pregnancy. Compared with normal-weight women, obese women have lower pregnancy rates (both spontaneous and by assisted reproductive technology), higher rates of early pregnancy loss and poorer oocyte quality. STUDY DESIGN, SIZE, DURATION: Beginning at 6 weeks of age, female C57Bl/6J mice were fed either a high-fat/high-sugar diet (HF/HS; 58% Fat Energy/Sucrose) or a diet of standard mouse chow (CON; 13% Fat) for 12 weeks. At this point, metabolic parameters were measured. Some of the mice (n = 9 HF/HS and 9 CON) were mated with reproductively competent males, and implantation sites were assessed. Other mice (n = 11 HF/HS and 10 CON) were mated with vasectomized males, and artificial decidualization was induced. For in vitro human studies of primary ESCs, endometrial tissue was obtained via biopsy from normo-ovulatory patients without history of infertility (obese = BMI > 30 kg/m(2), n = 11 and lean = BMI < 25 kg/m(2), n = 7) and from patients consented for hysterectomies for a benign indication (n = 4). In vitro studies were also performed with immortalized human ESCs. ESCs were decidualized in culture for nine 9 days in the presence or absence of palmitic acid (PA), and the degree of decidualization was assessed by measuring expression of decidualization markers. PARTICIPANTS/MATERIALS, SETTING, METHODS: The sizes of implantation sites and fetuses were analyzed in mice mated with reproductively competent males. In mice mated with vasectomized males, decidualization was induced, and uterine tissues were analyzed via hematoxylin and eosin staining, quantitative RT-PCR (RT-qPCR), and western blots. Human ESCs were cultured in vitro and induced to decidualize by treatment with cAMP and medroxyprogesterone. The level of expression of decidualization markers was assessed by RT-qPCR (mRNA) and western blotting (protein). ATP content of ESCs was measured, and levels of autophagy were assessed by western blotting of the autophagy regulators acetyl coa carboxylase (ACC) and ULK1 (Ser 317). Autophagic flux was measured by western blot of the marker LC3b-II. MAIN RESULTS AND THE ROLE OF CHANCE: Mice exposed to an HF/HS diet became obese and metabolically impaired. HF/HS-exposed mice mated to reproductively competent males had smaller implantation sites in early pregnancy (P <0.001) and larger fetuses at term (P <0.05) than CON-exposed mice. In the artificial decidualization experiments, mice exposed to the HF/HS diet developed 50% smaller deciduomas than mice exposed to CON diet (P< 0.001). Human ESCs cultured in the presence of PA had markedly decreased mRNA expression of the decidualization markers, decidual prolactin (PRL) (P< 0.0001) and insulin-like growth factor binding protein 1 (IGFBP1) (P< 0.0001). Expression of PRL and IGFBP1 by mRNA were also significantly lower in early follicular phase ESCs of obese women than in those of normal-weight women (P< 0.05). Protein expression of phosphorylated ACC and phosphorylated ULK1, both activated forms, were lower in deciduomas of HF/HS mice than in those of control mice (P < 0.01). In immortalized human ESCs, LC3b-II levels were higher in decidualized cells than in controls, indicating increased autophagy. PA treatment abrogated this increase. LIMITATIONS, REASONS FOR CAUTION: Many aspects of obesity and metabolic impairment could contribute to the decidualization defects observed in the HF/HS-exposed mice. Although our findings suggest that both autophagy and decidualization are impaired by exposure to PA, the underlying mechanisms should be elucidated. Finally, our human patient sample size was small. WIDER IMPLICATIONS OF THE FINDINGS: Although many factors contribute to poor reproductive outcome and early pregnancy loss in obese women, our study suggests the importance of decidualization defects. Such defects may contribute to compromised endometrial receptivity and poor implantation. If defects in autophagy contribute to impaired decidualization, therapeutics could be developed to improve this process and thus improve implantation and pregnancy outcomes in obese women. STUDY FUNDING/COMPETING INTERESTS: Grants include NIH 5T32HD040135-12 (J.S.R.), R01 HD065435 (K.H.M.), NIH T32 HD049305 (J.L.S.) and ACOG Research Grant (M.B.S.). The authors report no conflicts of interest.


Asunto(s)
Autofagia , Dieta Alta en Grasa , Obesidad/patología , Células del Estroma/patología , Animales , Biomarcadores/metabolismo , Decidua , Implantación del Embrión , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Ácido Palmítico/farmacología , Fosforilación , ARN Mensajero/metabolismo
2.
Reproduction ; 151(3): 261-70, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26700938

RESUMEN

Obesity negatively affects many aspects of the human body including reproductive function. In females, the root of the decline in fertility is linked to problems in the oocyte. Problems seen in oocytes that positively correlate with increasing BMI include changes to the metabolism, lipid accumulation, meiosis, and metaphase II (MII) spindle structure. Studies in mice indicate that dietary interventions fail to reverse these problems. How exercise affects the oocytes has not been addressed. Therefore, we hypothesized an exercise intervention would improve oocyte quality. Here we show that in a mouse model of an exercise, intervention can improve lipid metabolism in germinal vesicle (GV) stage oocytes. Oocytes significantly increased activity and transcription of the ß-oxidation enzyme hydroxyacyl-coenzyme A dehydrogenase in response to exercise training only if the mice had been fed a high-fat diet (HFD). An exercise intervention also reversed the lipid accumulation seen in GV stage oocytes of HFD females. However, delays in meiosis and disorganized MII spindles remained present. Therefore, exercise is able to improve, but not reverse, damage imparted on oocytes as a result of an HFD and obesity. By utilizing an exercise intervention on an HFD, we determined only lipid content, and lipid metabolism is changed in GV oocytes. Moving forward, interventions to improve oocyte quality may need to be more targeted to the oocyte specifically. Because of the HFD-induced deficiency in ß-oxidation, dietary supplementation with substrates to improve lipid utilization may be more beneficial.


Asunto(s)
Obesidad/terapia , Oocitos/metabolismo , Condicionamiento Físico Animal , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Mitocondrias/patología , Mitocondrias/ultraestructura , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Obesidad/metabolismo , Obesidad/patología , Huso Acromático/patología
3.
Reprod Fertil Dev ; 27(4): 716-24, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25775080

RESUMEN

Obesity adversely affects reproduction and results in oocyte defects in both mice and humans. In the present study we used a mouse model to examine whether the adverse effects of an obesogenic diet on oocyte metabolism and morphology can be reversed by return to a control diet. The intervention group consisted of C57BL6/J mice placed on a high-fat diet (HFD; 35.8% fat and 20.2% protein by nutritional content) for 6 weeks and then switched to an isocaloric control diet (CD; 13% fat and 25% protein) for 8 weeks (HFD/CD mice). The control group consisted of age-matched C57BL6/J mice maintained on CD for 14 weeks (CD/CD mice). Although metabolic parameters (weight, glucose tolerance and cholesterol levels) of HFD/CD mice returned to normal after this 'diet reversal' period, several oocyte defects were not reversible. These HFD/CD oocytes demonstrated significantly higher percentages of abnormal meiotic spindles, lower mitochondrial membrane potential and lower ATP and citrate levels, and higher percentages of abnormal lipid accumulation and mitochondrial distribution compared with CD/CD mice. These results suggest that the negative effects of an obesogenic diet on oocyte quality are not reversible, despite reversal of metabolic parameters. These data may provide better insight when counselling obese women regarding reproductive options and success.


Asunto(s)
Dieta Alta en Grasa , Obesidad/metabolismo , Oocitos/metabolismo , Animales , Peso Corporal/fisiología , Femenino , Ratones , Reproducción/fisiología
4.
Biol Reprod ; 90(2): 34, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24403548

RESUMEN

Embryo implantation and development requires the endometrial stromal cells (ESCs) to undergo decidualization. This differentiation process requires glucose utilization, and blockade of the pentose phosphate pathway inhibits decidualization of ESCs both in vitro and in vivo. Glucose and fatty acids are energy substrates for many cell types, and fatty acid beta-oxidation is critical for embryo implantation. Here, we investigated whether beta-oxidation is required for decidualization of ESCs. As assessed by marker gene expression, decidualization of human primary ESCs was blocked by reducing activity of carnitine calmitoyltransferase I, the rate-limiting enzyme in beta-oxidation, either by short hairpin RNA-mediated silencing or by treatment with the inhibitor etomoxir. Ranolazine (RAN), a partial beta-oxidation inhibitor, blocked early decidualization of a human ESC line. However, decidualization resumed after several days, most likely due to a compensatory up-regulation of GLUT1 expression and an increase in glucose metabolism. Simultaneous inhibition of the beta-oxidation pathway with RAN and the pentose phosphate pathway with glucosamine (GlcN) impaired in vitro decidualization of human ESCs more strongly than inhibition of either pathway alone. These findings were confirmed in murine ESCs in vitro, and exposure to RAN plus GlcN inhibited decidualization in vivo in a deciduoma model. Finally, intrauterine implantation of time-release RAN and GlcN pellets reduced pup number. Importantly, pup number returned to normal after the end of the pellet-active period. This work indicates that both fatty acids and glucose metabolism pathways are important for ESC decidualization, and suggests novel pathways to target for the design of future nonhormonal contraceptives.


Asunto(s)
Decidua/metabolismo , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos/fisiología , Células del Estroma/metabolismo , Animales , Células Cultivadas , Endometrio/citología , Endometrio/metabolismo , Femenino , Humanos , Masculino , Redes y Vías Metabólicas/fisiología , Ratones , Ratones Endogámicos ICR , Oxidación-Reducción
5.
Reprod Fertil Dev ; 27(1): 31-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25472042

RESUMEN

Obese women experience worse reproductive outcomes than normal weight women, specifically infertility, pregnancy loss, fetal malformations and developmental delay of offspring. The aim of the present study was to use a genetic mouse model of obesity to recapitulate the human reproductive phenotype and further examine potential mechanisms and therapies. New inbred, polygenic Type 2 diabetic TallyHO mice and age-matched control C57BL/6 mice were superovulated to obtain morula or blastocyst stage embryos that were cultured in human tubal fluid (HTF) medium. Deoxyglucose uptake was determined for individual insulin-stimulated blastocysts. Apoptosis was detected by confocal microscopy using the terminal deoxyribonucleotidyl transferase-mediated dUTP-digoxigenin nick end-labelling (TUNEL) assay and Topro-3 nuclear dye. Embryos were scored for TUNEL-positive as a percentage of total nuclei. AMP-activated protein kinase (AMPK) activation, tumour necrosis factor (TNF)-α expression and adiponectin expression were analysed by western immunoblot and confocal immunofluorescent microscopy. Lipid accumulation was assayed by BODIPY. Comparisons were made between TallyHO morulae cultured to blastocyst embryos in either HTF medium or HTF medium with 25 µg mL(-1) metformin. TallyHO mice developed whole body abnormal insulin tolerance, had decreased litter sizes and increased non-esterified fatty acid levels. Blastocysts from TallyHO mice exhibited increased apoptosis, decreased insulin sensitivity and decreased AMPK. A possible cause for the insulin resistance and abnormal AMPK phosphorylation was the increased TNF-α expression and lipid accumulation, as detected by BODIPY, in TallyHO blastocysts and decreased adiponectin. Culturing TallyHO morulae with the AMPK activator metformin led to a reversal of all the abnormal findings, including increased AMPK phosphorylation, improved insulin-stimulated glucose uptake and normalisation of lipid accumulation. Women with obesity and insulin resistance experience poor pregnancy outcomes. Previously we have shown in mouse models of insulin resistance that AMPK activity is decreased and that activators of AMPK reverse poor embryo outcomes. Here, we show for the first time using a genetically altered obese model, not a diet-induced model, that metformin reverses many of the adverse effects of obesity at the level of the blastocyst. Expanding on this we determine that activation of AMPK via metformin reduces lipid droplet accumulation, presumably by eliminating the inhibitory effects of TNF-α, resulting in normalisation of fatty acid oxidation and HADH2 (hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase (trifunctional protein), alpha subunit) activity. Metformin exposure in vitro was able to partially reverse these effects, at the level of the blastocyst, and may thus be effective in preventing the adverse effects of obesity on pregnancy and reproductive outcomes.


Asunto(s)
Blastocisto/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Metformina/farmacología , Obesidad/etiología , Reproducción/efectos de los fármacos , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Ácidos Grasos/metabolismo , Femenino , Humanos , Etiquetado Corte-Fin in Situ , Resistencia a la Insulina/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Microscopía Confocal , Embarazo
6.
Biol Reprod ; 89(1): 16, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23718985

RESUMEN

Embryo implantation in the uterus depends on decidualization of the endometrial stromal cells (ESCs), and glucose utilization via the pentose phosphate pathway is critical in this process. We hypothesized that the amino sugar glucosamine may block the pentose phosphate pathway via inhibition of the rate-limiting enzyme glucose-6-phosphate dehydrogenase in ESCs and therefore impair decidualization and embryo implantation, thus preventing pregnancy. Both human primary and immortalized ESCs were decidualized in vitro in the presence of 0, 2.5, or 5 mM glucosamine for 9 days. Viability assays demonstrated that glucosamine was well tolerated by human ESCs. Exposure of human ESCs to glucosamine resulted in significant decreases in the activity and expression of glucose-6-phosphate dehydrogenase and in the mRNA expression of the decidual markers prolactin, somatostatin, interleukin-15, and left-right determination factor 2. In mouse ESCs, expression of the decidual marker Prp decreased upon addition of glucosamine. In comparison with control mice, glucosamine-treated mice showed weak artificial deciduoma formation along the stimulated uterine horn. In a complementary in vivo experiment, a 60-day-release glucosamine (15, 150, or 1500 µg) or placebo pellet was implanted in a single uterine horn of mice. Mice with a glucosamine pellet delivered fewer live pups per litter than those with a control pellet, and pup number returned to normal after the end of the pellet-active period. In conclusion, glucosamine is a nonhormonal inhibitor of decidualization of both human and mouse ESCs and of pregnancy in mice. Our data indicate the potential for development of glucosamine as a novel, reversible, nonhormonal contraceptive.


Asunto(s)
Endometrio/efectos de los fármacos , Glucosamina/farmacología , Tamaño de la Camada/efectos de los fármacos , Animales , Anticonceptivos , Endometrio/citología , Endometrio/enzimología , Femenino , Glucosafosfato Deshidrogenasa/metabolismo , Humanos , Ratones , Embarazo , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología
7.
Am J Physiol Endocrinol Metab ; 302(12): E1511-8, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22473000

RESUMEN

Glucose is an essential nutrient for mammalian cells. Emerging evidence suggests that glucose within the oocyte regulates meiotic maturation. However, it remains controversial as to whether, and if so how, glucose enters oocytes within cumulus-oocyte complexes (COCs). We used a fluorescent glucose derivative (6-NBDG) to trace glucose transport within live mouse COCs and employed inhibitors of glucose transporters (GLUTs) and gap junction proteins to examine their distinct roles in glucose uptake by cumulus cells and the oocyte. We showed that fluorescent glucose enters both cumulus-enclosed and denuded oocytes. Treating COCs with GLUT inhibitors leads to simultaneous decreases in glucose uptake in cumulus cells and the surrounded oocyte but no effect on denuded oocytes. Pharmacological blockade of of gap junctions between the oocyte and cumulus cells significantly inhibited fluorescent glucose transport to oocytes. Moreover, we find that both in vivo hyperglycemic environment and in vitro high-glucose culture increase free glucose levels in oocytes via gap junctional channels. These findings reveal an intercellular pathway for glucose transport into oocytes: glucose is taken up by cumulus cells via the GLUT system and then transferred into the oocyte through gap junctions. This intercellular pathway may partly mediate the effects of high-glucose condition on oocyte quality.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Oocitos/metabolismo , 4-Cloro-7-nitrobenzofurazano/análogos & derivados , Animales , Núcleo Celular/metabolismo , Conexinas/metabolismo , Células del Cúmulo/metabolismo , Femenino , Colorantes Fluorescentes , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Glucosamina/análogos & derivados , Glucosa/metabolismo , Glucosa/farmacología , Proteínas Facilitadoras del Transporte de la Glucosa/antagonistas & inhibidores , Ratones , Ratones Endogámicos ICR , Oocitos/enzimología , Zona Pelúcida/metabolismo
8.
Biol Reprod ; 87(2): 49, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22649075

RESUMEN

SLC2A8, also known as GLUT8, is a facilitative glucose transporter expressed in the testis, brain, liver, heart, uterus, ovary, and fat. In this study we examined the effect of Slc2a8 deficiency on mouse gamete, preimplantation embryo, and implantation phenotype, as well as postnatal growth and physiology. For this model, the transcriptional start site and exons 1-4 were targeted and a lack of protein expression was confirmed by Western immunoblot. Oocytes obtained from Slc2a8(-/-) mice demonstrated abnormal metabolism and ATP production. In addition, deletion of Slc2a8 resulted in impaired decidualization, a critical step in the differentiation of endometrial stromal cells (ESCs), necessary for implantation. This indicates a role for SLC2A8 in decidualization, which is supported by Slc2a8 mRNA expression in both mouse and human ESCs, which increases dramatically in response to hormonal changes occurring during the process of implantation. Ovarian transplantation studies confirm that lack of SLC2A8 affects both the embryo and the implantation processes. This phenotype leads to decreased litter size, and smaller pups at weaning that continue to display an abnormally small growth phenotype into adulthood. The Slc2a8 null mice display decreased body fat by magnetic resonance imaging, and, interestingly, they are resistant to a diet high in fat and carbohydrates.


Asunto(s)
Decidua/fisiología , Implantación del Embrión , Proteínas Facilitadoras del Transporte de la Glucosa/fisiología , Animales , Peso Corporal , Dieta Alta en Grasa , Femenino , Glucosa/metabolismo , Homeostasis , Tamaño de la Camada , Masculino , Ratones , Ratones Noqueados , Oocitos/metabolismo , Ovario/anatomía & histología , Fenotipo , Motilidad Espermática , Testículo/anatomía & histología , Útero/fisiología
9.
Biol Reprod ; 85(4): 678-83, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21653893

RESUMEN

Free fatty acids (FFAs) are energy substrates for many cell types, but in excess, some FFAs can accumulate in nonadipose cells, inducing apoptosis. Also known as lipotoxicity, this phenomenon may play a role in the development of obesity-related disease. Obesity is common among reproductive age women and is associated with adverse pregnancy and fetal outcomes; however, little is known about the effects of excess FFAs on embryos and subsequent fetal development. To address this knowledge gap, murine blastocysts were cultured in excess palmitic acid (PA), the most abundant saturated FFA in human serum, and ovarian follicular fluid. Targets susceptible to aberrations in maternal physiology, including embryonic IGF1 receptor (IGF1R) expression, glutamic pyruvate transaminase (GPT2) activity, and nuclei count, were measured. PA-exposed blastocysts demonstrated altered IGF1R expression, increased GPT2 activity, and decreased nuclei count. Trophoblast stem cells derived from preimplantation embryos were also cultured in PA. Cells exposed to increasing doses of PA demonstrated increased apoptosis and decreased proliferation. To demonstrate long-term effects of brief PA exposure, blastocysts cultured for 30 h in PA were transferred into foster mice, and pregnancies followed through Embryonic Day (ED)14.5 or delivery. Fetuses resulting from PA-exposed blastocysts were smaller than controls at ED14.5. Delivered pups were also smaller but demonstrated catch-up growth and ultimately surpassed control pups in weight. Altogether, our data suggest brief PA exposure results in altered embryonic metabolism and growth, with lasting adverse effects on offspring, providing further insight into the pathophysiology of maternal obesity.


Asunto(s)
Blastocisto/metabolismo , Desarrollo Fetal , Retardo del Crecimiento Fetal/etiología , Obesidad/etiología , Ácido Palmítico/efectos adversos , Animales , Apoptosis , Blastocisto/citología , Peso Corporal , Recuento de Células , Proliferación Celular , Células Cultivadas , Cruzamientos Genéticos , Ectogénesis , Transferencia de Embrión , Femenino , Feto/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Embarazo , Transaminasas/metabolismo , Trofoblastos/citología , Trofoblastos/metabolismo
10.
Reproduction ; 141(5): 607-15, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21367963

RESUMEN

Autophagy is critical to the process of development because mouse models have shown that lack of autophagy leads to developmental arrest during the pre-implantation stage of embryogenesis. The process of autophagy is regulated through signaling pathways, which respond to the cellular environment. Therefore, any alteration in the environment may lead to the dysregulation of the autophagic process potentially resulting in cell death. Using both in vitro and in vivo models to study autophagy in the pre-implantation murine embryo, we observed that the cells respond to environmental stressors (i.e. hyperglycemic environment) by increasing activation of autophagy in a differential pattern within the embryo. This upregulation is accompanied by an increase in apoptosis, which appears to plateau at high concentrations of glucose. The activation of the autophagic pathway was further confirmed by an increase in GAPDH activity in both in vivo and in vitro hyperglycemic models, which has been linked to autophagy through the activation of the Atg12 gene. Furthermore, this increase in autophagy in response to a hyperglycemic environment was observed as early as the oocyte stage. In conclusion, in this study, we provided evidence for a differential response of elevated activation of autophagy in embryos and oocytes exposed to a hyperglycemic environment.


Asunto(s)
Autofagia , Blastocisto/metabolismo , Glucosa/metabolismo , Hiperglucemia/metabolismo , Transducción de Señal , Estrés Fisiológico , Animales , Blastocisto/patología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Técnicas de Cultivo de Embriones , Desarrollo Embrionario , Femenino , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Hiperglucemia/embriología , Hiperglucemia/patología , Ratones , Oocitos/metabolismo , Oocitos/ultraestructura , Regulación hacia Arriba
11.
Reproduction ; 138(5): 759-70, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19700529

RESUMEN

cAMP plays a critical role in the control of oocyte maturation, as a high level of cAMP maintains oocyte arrest at the first meiotic prophase. Yet this study shows that pulsing meiotically arrested denuded oocytes (DO) with cAMP induces oocyte maturation through the activation of AMP-activated protein kinase (PRKA). Short-term (3 h) pulsing of meiotically arrested oocytes with forskolin, an adenyl cyclase (AC) activator, increased oocyte cAMP, led to elevated AMP, and induced oocyte meiotic resumption compared to oocytes continuously cultured in the control medium with or without forskolin. Western analysis showed that germinal vesicle (GV)-stage oocytes after forskolin pulsing contained increased levels of phospho-acetyl CoA carboxylase (pACACA), a primary substrate of PRKA. Pulsing oocytes with the phosphodiesterase (PDE)-sensitive cAMP analog, 8-bromo-cAMP (8-Br-cAMP), also increased pACACA and pPRKA levels in GV-stage oocytes and induced oocyte meiotic resumption. Moreover, the PRKA inhibitors, compound C and araA, prevented 8-Br-cAMP pulsing-induced maturation. The lack of effect on meiotic induction and PRKA activation when oocytes were pulsed with the PDE-resistant activators of cAMP-dependent protein kinase, Sp-cAMP-AM and Sp-5,6-DCI-cBIMPS, suggests that cAMP degradation is required for pulsing-induced maturation. Pulsing oocytes with the exchange protein directly activated by cAMP (Epac)-specific activator, 8-CPT-2'-O-Me-cAMP, had no stimulatory effect on oocyte maturation, suggesting Epac is not involved in the pulsing-induced maturation. Taken together, these data support the idea that a transient increase in oocyte cAMP can induce meiotic resumption via activation of PRKA.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , AMP Cíclico/farmacología , Meiosis/efectos de los fármacos , Oocitos/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/fisiología , Animales , Células Cultivadas , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Femenino , Meiosis/fisiología , Ratones , Ratones Endogámicos C57BL , Oocitos/citología , Oocitos/metabolismo , Oocitos/fisiología , Oogénesis/efectos de los fármacos , Oogénesis/fisiología , Periodicidad , Regulación hacia Arriba/efectos de los fármacos , Zona Pelúcida
12.
Reproduction ; 136(3): 335-44, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18577554

RESUMEN

Maternal insulin resistance results in poor pregnancy outcomes. In vivo and in vitro exposure of the murine blastocyst to high insulin or IGF1 results in the down-regulation of the IGF1 receptor (IGF1R). This in turn leads to decreased glucose uptake, increased apoptosis, as well as pregnancy resorption and growth restriction. Recent studies have shown that blastocyst activation of AMP-activated protein kinase (AMPK) reverses these detrimental effects; however, the mechanism was not clear. The objective of this study was to determine how AMPK activation rescues the insulin-resistant blastocyst. Using trophoblast stem (TS) cells derived from the blastocyst, insulin resistance was recreated by transfecting with siRNA to Igf1r and down-regulating expression of the protein. These cells were then exposed to AMPK activators 5-aminoimidazole-4-carboxamide riboside and phenformin, and evaluated for apoptosis, insulin-stimulated 2-deoxyglucose uptake, PI3-kinase activity, and levels of phospho-AKT, phospho-mTor, and phospho-70S6K. Surprisingly, disrupted insulin signaling led to decreased AMPK activity in TS cells. Activators reversed these effects by increasing the AMP/ATP ratio. Moreover, this treatment increased insulin-stimulated 2-deoxyglucose transport and cell survival, and led to an increase in PI3-kinase activity, as well as increased P-mTOR and p70S6K levels. This study is the first to demonstrate significant crosstalk between the AMPK and insulin signaling pathways in embryonic cells, specifically the enhanced response of PI3K/AKT/mTOR to AMPK activation. Decreased insulin signaling also resulted in decreased AMPK activation. These findings provide mechanistic targets in the AMPK signaling pathway that may be essential for improved pregnancy success in insulin-resistant states.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Blastocisto/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Transducción de Señal/fisiología , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting/métodos , Línea Celular , Desoxiglucosa/metabolismo , Activación Enzimática , Femenino , Hipoglucemiantes/farmacología , Ratones , Fenformina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Receptor IGF Tipo 1/genética , Ribonucleósidos/farmacología , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo
13.
Sci Rep ; 6: 25091, 2016 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-27125896

RESUMEN

Maternal metabolic diseases increase offspring risk for low birth weight and cardiometabolic diseases in adulthood. Excess fructose consumption may confer metabolic risks for both women and their offspring. However, the direct consequences of fructose intake per se are unknown. We assessed the impact of a maternal high-fructose diet on the fetal-placental unit in mice in the absence of metabolic syndrome and determined the association between maternal serum fructose and placental uric acid levels in humans. In mice, maternal fructose consumption led to placental inefficiency, fetal growth restriction, elevated fetal serum glucose and triglyceride levels. In the placenta, fructose induced de novo uric acid synthesis by activating the activities of the enzymes AMP deaminase and xanthine oxidase. Moreover, the placentas had increased lipids and altered expression of genes that control oxidative stress. Treatment of mothers with the xanthine oxidase inhibitor allopurinol reduced placental uric acid levels, prevented placental inefficiency, and improved fetal weights and serum triglycerides. Finally, in 18 women delivering at term, maternal serum fructose levels significantly correlated with placental uric acid levels. These findings suggest that in mice, excess maternal fructose consumption impairs placental function via a xanthine oxidase/uric acid-dependent mechanism, and similar effects may occur in humans.


Asunto(s)
Retardo del Crecimiento Fetal/inducido químicamente , Fructosa/sangre , Placenta/metabolismo , Insuficiencia Placentaria/inducido químicamente , Ácido Úrico/metabolismo , AMP Desaminasa/metabolismo , Alopurinol/administración & dosificación , Alopurinol/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/prevención & control , Fructosa/efectos adversos , Ratones , Estrés Oxidativo , Insuficiencia Placentaria/prevención & control , Embarazo , Triglicéridos/sangre , Xantina Oxidasa/antagonistas & inhibidores , Xantina Oxidasa/metabolismo
14.
Cell Rep ; 16(1): 1-8, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27320925

RESUMEN

Maternal obesity impairs offspring health, but the responsible mechanisms are not fully established. To address this question, we fed female mice a high-fat/high-sugar diet from before conception until weaning and then followed the outcomes in the next three generations of offspring, all fed a control diet. We observed that female offspring born to obese mothers had impaired peripheral insulin signaling that was associated with mitochondrial dysfunction and altered mitochondrial dynamic and complex proteins in skeletal muscle. This mitochondrial phenotype persisted through the female germline and was passed down to the second and third generations. Our results indicate that maternal programming of metabolic disease can be passed through the female germline and that the transfer of aberrant oocyte mitochondria to subsequent generations may contribute to the increased risk for developing insulin resistance.


Asunto(s)
Células Germinativas/metabolismo , Síndrome Metabólico/metabolismo , Mitocondrias/patología , Animales , Dieta Alta en Grasa , Carbohidratos de la Dieta , Femenino , Intolerancia a la Glucosa/complicaciones , Patrón de Herencia , Resistencia a la Insulina , Masculino , Síndrome Metabólico/complicaciones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Dinámicas Mitocondriales , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Oocitos/metabolismo , Oocitos/ultraestructura , Embarazo , Transducción de Señal
15.
Sci Signal ; 9(416): ra21, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26905426

RESUMEN

Trehalose is a naturally occurring disaccharide that has gained attention for its ability to induce cellular autophagy and mitigate diseases related to pathological protein aggregation. Despite decades of ubiquitous use as a nutraceutical, preservative, and humectant, its mechanism of action remains elusive. We showed that trehalose inhibited members of the SLC2A (also known as GLUT) family of glucose transporters. Trehalose-mediated inhibition of glucose transport induced AMPK (adenosine 5'-monophosphate-activated protein kinase)-dependent autophagy and regression of hepatic steatosis in vivo and a reduction in the accumulation of lipid droplets in primary murine hepatocyte cultures. Our data indicated that trehalose triggers beneficial cellular autophagy by inhibiting glucose transport.


Asunto(s)
Autofagia , Hígado Graso/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Trehalosa/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Hígado Graso/genética , Hígado Graso/patología , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Células HEK293 , Células Hep G2 , Humanos , Ratones , Ratones Noqueados
16.
Mol Endocrinol ; 17(10): 2096-102, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12829803

RESUMEN

Target-soluble N-ethylmaleimide-sensitive factor attachment protein receptors (t-SNAREs) are receptors that facilitate vesicle and target membrane fusion. Syntaxin 4 is the t-SNARE critical for insulin-stimulated glucose transporter 4 (GLUT4)-plasma membrane fusion in adipocytes. GLUT8 is a novel IGF-I/insulin-regulated glucose transporter expressed in the mouse blastocyst. Similar to GLUT4, GLUT8 translocates to the plasma membrane to increase glucose uptake at a stage in development when glucose serves as the main substrate. Any decrease in GLUT8 cell surface expression results in increased apoptosis and pregnancy loss. Previous studies have also shown that disruption of the syntaxin 4 (Stx4a) gene results in early embryonic lethality before embryonic d 7.5. We have now demonstrated that syntaxin 4 protein is localized predominantly to the apical plasma membrane of the murine blastocyst. Stx4a inheritance, as detected by protein expression, occurs with the expected Mendelian frequency up to embryonic d 4.5. In parallel, 22% of the blastocysts from Stx4a+/- matings had no significant insulin-stimulated translocation of GLUT8 whereas 77% displayed either partial or complete translocation to the apical plasma membrane. This difference in GLUT8 translocation directly correlated with one-third of blastocysts from Stx4a+/- mating having reduced rates of insulin-stimulated glucose uptake and 67% with wild-type rates. These data demonstrate that the lack of syntaxin 4 expression results in abnormal movement of GLUT8 in response to insulin, decreased insulin-stimulated glucose uptake, and increased apoptosis. Thus, syntaxin 4 functions as the necessary t-SNARE protein responsible for correct fusion of the GLUT8-containing vesicle with the plasma membrane in the mouse blastocyst.


Asunto(s)
Blastocisto/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Animales , Apoptosis , Femenino , Expresión Génica , Proteínas Facilitadoras del Transporte de la Glucosa , Insulina/farmacología , Masculino , Proteínas de la Membrana/genética , Ratones , Embarazo , Transporte de Proteínas , Proteínas Qa-SNARE , Proteínas R-SNARE , Proteína 3 de Membrana Asociada a Vesículas
17.
Reprod Sci ; 22(1): 94-101, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24803506

RESUMEN

We set out to determine whether the addition of an aryl hydrocarbon receptor (AHR) antagonist has an effect on glucose/fructose utilization in the spermatocyte when exposed to cigarette smoke condensate (CSC). We exposed male germ cells to 5 and 40 µg/mL of CSC ± 10 µmol/L of AHR antagonist at various time points. Immunoblot expression of specific glucose/fructose transporters was compared to control. Radiolabeled uptake of 2-deoxyglucose (2-DG) and fructose was also performed. Spermatocytes utilized fructose nearly 50-fold more than 2-DG. Uptake of 2-DG decreased after CSC + AHR antagonist exposure. Glucose transporters (GLUTs) 9a and 12 declined after CSC + AHR antagonist exposure. Synergy between CSC and the AHR antagonist in spermatocytes may disrupt the metabolic profile in vitro. Toxic exposures alter energy homeostasis in early stages of male germ cell development, which could contribute to later effects explaining decreases in sperm motility in smokers.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Hexosas/metabolismo , Humo/efectos adversos , Fumar/efectos adversos , Espermatocitos/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Compuestos Azo/toxicidad , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular , Desoxiglucosa/metabolismo , Fructosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Masculino , Ratones , Pirazoles/toxicidad , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/metabolismo , Espermatocitos/metabolismo , Factores de Tiempo
18.
Aging Cell ; 14(5): 887-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26111777

RESUMEN

Female reproductive capacity declines dramatically in the fourth decade of life as a result of an age-related decrease in oocyte quality and quantity. The primary causes of reproductive aging and the molecular factors responsible for decreased oocyte quality remain elusive. Here, we show that aging of the female germ line is accompanied by mitochondrial dysfunction associated with decreased oxidative phosphorylation and reduced Adenosine tri-phosphate (ATP) level. Diminished expression of the enzymes responsible for CoQ production, Pdss2 and Coq6, was observed in oocytes of older females in both mouse and human. The age-related decline in oocyte quality and quantity could be reversed by the administration of CoQ10. Oocyte-specific disruption of Pdss2 recapitulated many of the mitochondrial and reproductive phenotypes observed in the old females including reduced ATP production and increased meiotic spindle abnormalities, resulting in infertility. Ovarian reserve in the oocyte-specific Pdss2-deficient animals was diminished, leading to premature ovarian failure which could be prevented by maternal dietary administration of CoQ10. We conclude that impaired mitochondrial performance created by suboptimal CoQ10 availability can drive age-associated oocyte deficits causing infertility.


Asunto(s)
Envejecimiento/efectos de los fármacos , Fertilidad/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Oocitos/efectos de los fármacos , Reproducción/efectos de los fármacos , Ubiquinona/análogos & derivados , Animales , Femenino , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Oocitos/citología , Oocitos/metabolismo , Ubiquinona/farmacología
19.
Endocrinology ; 145(3): 1435-43, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14657010

RESUMEN

During preimplantation development in the mouse, it is crucial that glucose metabolism not be compromised. Any decrease in glucose uptake at this stage in development can compromise the developing embryo. We have cloned another member of the glucose transporter family, GLUT9, which is expressed embryonically. Three different isoforms were identified. We have shown that two of the mouse GLUT9 isoforms transport glucose at a rate significantly greater than controls. Expression analysis of the preimplantation blastocyst identifies only the presence of the shorter GLUT9 isoform, RT-PCR and Western immunoblot confirmed this finding. A differential pattern of expression was seen with GLUT9 present at the plasma membrane in one- and two-cell zygotes and in an intracellular compartment in trophectoderm cells at a blastocyst stage. Although blocking GLUT9 expression during preimplantation development had no effect on glucose transport or apoptosis, transfer of these embryos into pseudopregnant mice resulted in increased pregnancy loss, suggesting that GLUT9 is critical for early preimplantation development.


Asunto(s)
Blastocisto/fisiología , Proteínas de Transporte de Monosacáridos/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Glucosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/metabolismo
20.
Endocrinology ; 155(8): 2781-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24840347

RESUMEN

Type 1 diabetes is associated with subfertility in humans. The current treatment for type 1 diabetes, insulin monotherapy, is suboptimal to fully stabilize glycemia, potentially leading to this subfertility. Recent work has demonstrated that treatment with the energy-regulating hormone leptin, alone or in combination with insulin, can more effectively control glycemia in mouse models of type 1 diabetes. Here, we sought to determine whether the fertility defects in a type 1 diabetic mouse model, the Akita mouse, can be rescued with leptin monotherapy in the absence of any exogenous insulin. Akita homozygous mice treated with leptin alone had a larger total body size, testes, and seminal vesicles than their untreated siblings. Leptin treatment prevented testicular degeneration and rescued sperm motility to wild-type levels. Furthermore, sperm obtained from leptin-treated mice could successfully fertilize ooctyes in vitro. Despite completely rescuing spermatogenesis, the critical reproductive hormones LH and testosterone were only modestly higher than in untreated mice, indicating that a minimum threshold of these hormones must be met to maintain spermatogenesis. Cumulatively, these findings implicate the importance of leptin in maintaining fertility and support the use of leptin therapy in the treatment of type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/terapia , Infertilidad Masculina/terapia , Leptina/fisiología , Espermatogénesis/genética , Adiposidad/genética , Animales , Atrofia/genética , Atrofia/prevención & control , Atrofia/terapia , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Tamización de Portadores Genéticos , Homocigoto , Infertilidad Masculina/etiología , Infertilidad Masculina/patología , Leptina/genética , Leptina/uso terapéutico , Hormona Luteinizante/genética , Masculino , Ratones , Testosterona/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA