Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Int J Mol Sci ; 24(21)2023 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-37958743

RESUMO

Fetuses with intrauterine growth restriction (FGR) have impaired oxidative and energy metabolism, with persistent consequences on their postnatal development. In this study, we test the hypothesis that FGR skeletal muscle has lower mitochondrial respiration rate and alters the transcriptomic profiles associated with energy metabolism in an ovine model. At late gestation, mitochondrial oxygen consumption rates (OCRs) and transcriptome profiles were evaluated in the skeletal muscle collected from FGR and control fetuses. The ex vivo mitochondrial OCRs were reduced (p < 0.01) in permeabilized FGR soleus muscle compared to the control muscle but only with pyruvate as the metabolic substrate. Mitochondrial OCRs were similar between the FGR and control groups for palmitoyl-carnitine (fatty acid-driven) or pyruvate plus palmitoyl-carnitine metabolic substrates. A total of 2284 genes were differentially expressed in the semitendinosus muscle from growth restricted fetuses (false discovery rate (FDR) ≤ 0.05). A pathway analysis showed that the upregulated genes (FGR compared to control) were overrepresented for autophagy, HIF-1, AMPK, and FOXO signaling pathways (all with an FDR < 0.05). In addition, the expression of genes modulating pyruvate's entry into the TCA cycle was downregulated, whereas the genes encoding key fatty acid oxidation enzymes were upregulated in the FGR muscle. These findings show that FGR skeletal muscle had attenuated mitochondrial pyruvate oxidation, possibly associated with the inability of pyruvate to enter into the TCA cycle, and that fatty acid oxidation might compensate for the attenuated energy metabolism. The current study provided phenotypic and molecular evidence for adaptive deficiencies in FGR skeletal muscle.


Assuntos
Ácidos Graxos , Ácido Pirúvico , Feminino , Humanos , Animais , Ovinos , Gravidez , Ácidos Graxos/metabolismo , Ácido Pirúvico/metabolismo , Músculo Esquelético/metabolismo , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/metabolismo , Feto/metabolismo , Respiração , Palmitoilcarnitina
2.
Am J Physiol Endocrinol Metab ; 323(4): E336-E353, 2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-35858246

RESUMO

Infants born to obese mothers are more likely to develop metabolic disease, including glucose intolerance and hepatic steatosis, in adult life. We examined the effects of maternal obesity on the transcriptome of skeletal muscle and liver tissues of the near-term fetus and 3-mo-old offspring in mice born to dams fed a high-fat and -sugar diet. Previously, we have shown that male, but not female, offspring develop glucose intolerance, insulin resistance, and liver steatosis at 3 mo old. Female C57BL6/J mice were fed normal chow or an obesogenic high-calorie diet before mating and throughout pregnancy. RNAseq was performed on the liver and gastrocnemius muscle following collection from fetuses on embryonic day 18.5 (E18.5) as well as from 3-mo-old offspring from obese dams and control dams. Significant genes were generated for each sex, queried for enrichment, and modeled to canonical pathways. RNAseq was corroborated by protein quantification in offspring. The transcriptomic response to maternal obesity in the liver was more marked in males than females. However, in both male and female offspring of obese dams, we found significant enrichment for fatty acid metabolism, mitochondrial transport, and oxidative stress in the liver transcriptomes as well as decreased protein concentrations of electron transport chain members. In skeletal muscle, pathway analysis of gene expression revealed sexual dimorphic patterns, including metabolic processes of fatty acids and glucose, as well as PPAR, AMPK, and PI3K-Akt signaling pathways. Transcriptomic responses to maternal obesity in skeletal muscle were more marked in female offspring than males. Female offspring had greater expression of genes associated with glucose uptake, and protein abundance reflected greater activation of mTOR signaling. Skeletal muscle and livers in mice born to obese dams had sexually dimorphic transcriptomic responses that changed from the fetus to the adult offspring. These data provide insights into mechanisms underpinning metabolic programming in maternal obesity.NEW & NOTEWORTHY Transcriptomic data support that fetuses of obese mothers modulate metabolism in both muscle and liver. These changes were strikingly sexually dimorphic in agreement with published findings that male offspring of obese dams exhibit pronounced metabolic disease earlier. In both males and females, the transcriptomic responses in the fetus were different than those at 3 mo, implicating adaptive mechanisms throughout adulthood.


Assuntos
Fígado Gorduroso , Intolerância à Glucose , Obesidade Materna , Efeitos Tardios da Exposição Pré-Natal , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Dieta Hiperlipídica , Ácidos Graxos/metabolismo , Fígado Gorduroso/metabolismo , Feminino , Glucose/metabolismo , Intolerância à Glucose/metabolismo , Humanos , Insulina/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Obesos , Músculo Esquelético/metabolismo , Obesidade/genética , Obesidade/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/genética , Fosfatidilinositol 3-Quinases/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Transcriptoma
3.
Am J Physiol Endocrinol Metab ; 319(1): E67-E80, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32396498

RESUMO

Fetal sheep with placental insufficiency-induced intrauterine growth restriction (IUGR) have lower hindlimb oxygen consumption rates (OCRs), indicating depressed mitochondrial oxidative phosphorylation capacity in their skeletal muscle. We hypothesized that OCRs are lower in skeletal muscle mitochondria from IUGR fetuses, due to reduced electron transport chain (ETC) activity and lower abundances of tricarboxylic acid (TCA) cycle enzymes. IUGR sheep fetuses (n = 12) were created with mid-gestation maternal hyperthermia and compared with control fetuses (n = 12). At 132 ± 1 days of gestation, biceps femoris muscles were collected, and the mitochondria were isolated. Mitochondria from IUGR muscle have 47% lower State 3 (Complex I-dependent) OCRs than controls, whereas State 4 (proton leak) OCRs were not different between groups. Furthermore, Complex I, but not Complex II or IV, enzymatic activity was lower in IUGR fetuses compared with controls. Proteomic analysis (n = 6/group) identified 160 differentially expressed proteins between groups, with 107 upregulated and 53 downregulated mitochondria proteins in IUGR fetuses compared with controls. Although no differences were identified in ETC subunit protein abundances, abundances of key TCA cycle enzymes [isocitrate dehydrogenase (NAD+) 3 noncatalytic subunit ß (IDH3B), succinate-CoA ligase ADP-forming subunit-ß (SUCLA2), and oxoglutarate dehydrogenase (OGDH)] were lower in IUGR mitochondria. IUGR mitochondria had a greater abundance of a hypoxia-inducible protein, NADH dehydrogenase 1α subcomplex 4-like 2, which is known to incorporate into Complex I and lower Complex I-mediated NADH oxidation. Our findings show that mitochondria from IUGR skeletal muscle adapt to hypoxemia and hypoglycemia by lowering Complex I activity and TCA cycle enzyme concentrations, which together, act to lower OCR and NADH production/oxidation in IUGR skeletal muscle.


Assuntos
Ciclo do Ácido Cítrico/fisiologia , Complexo I de Transporte de Elétrons/metabolismo , Retardo do Crescimento Fetal/metabolismo , Mitocôndrias Musculares/metabolismo , Animais , Regulação para Baixo , Complexo II de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Retardo do Crescimento Fetal/enzimologia , Músculos Isquiossurais/enzimologia , Músculos Isquiossurais/metabolismo , Hipoglicemia/enzimologia , Hipoglicemia/metabolismo , Hipóxia/enzimologia , Hipóxia/metabolismo , Isocitrato Desidrogenase/metabolismo , Complexo Cetoglutarato Desidrogenase/metabolismo , Mitocôndrias Musculares/enzimologia , Proteínas Mitocondriais/metabolismo , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Consumo de Oxigênio , Insuficiência Placentária/enzimologia , Insuficiência Placentária/metabolismo , Gravidez , Proteômica , Ovinos , Succinato-CoA Ligases/metabolismo , Regulação para Cima
4.
Am J Physiol Regul Integr Comp Physiol ; 319(3): R255-R263, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32667834

RESUMO

Fetal conditions associated with placental insufficiency and intrauterine growth restriction (IUGR) chronically elevate plasma norepinephrine (NE) concentrations. Our objective was to evaluate the effects of chronically elevated NE on insulin-stimulated glucose metabolism in normally grown, non-IUGR fetal sheep, which are independent of other IUGR-related reductions in nutrients and oxygen availability. After surgical placement of catheters, near-term fetuses received either a saline (control) or NE intravenous infusion with controlled euglycemia. In NE fetuses, plasma NE concentrations were 5.5-fold greater than controls, and fetal euglycemia was maintained with a maternal insulin infusion. Insulin secretion was blunted in NE fetuses during an intravenous glucose tolerance test. Weight-specific fluxes for glucose were measured during a euinsulinemic-euglycemic clamp (EEC) and a hyperinsulinemic-euglycemic clamp (HEC). Plasma glucose and insulin concentrations were not different between groups within each clamp, but insulin concentrations increased 10-fold between the EEC and the HEC. During the EEC, rates of glucose uptake (umbilical uptake + exogenous infusion) and glucose utilization were 47% and 35% lower (P < 0.05) in NE fetuses compared with controls. During the HEC, rates of glucose uptake were 28% lower (P < 0.05) in NE fetuses than controls. Glucose production was undetectable in either group, and glucose oxidation was unaffected by the NE infusion. These findings indicate that chronic exposure to high plasma NE concentrations lowers rates of net glucose uptake in the fetus without affecting glucose oxidation rates or initiating endogenous glucose production. Lower fetal glucose uptake was independent of insulin, which indicates insulin resistance as a consequence of chronically elevated NE.


Assuntos
Glicemia/metabolismo , Feto/metabolismo , Norepinefrina/sangue , Insuficiência Placentária/metabolismo , Animais , Feminino , Retardo do Crescimento Fetal/metabolismo , Insulina/sangue , Resistência à Insulina/fisiologia , Ilhotas Pancreáticas/metabolismo , Gravidez , Ovinos
5.
Clin Sci (Lond) ; 134(8): 961-984, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32313958

RESUMO

Maternal obesity is associated with pregnancy complications and increases the risk for the infant to develop obesity, diabetes and cardiovascular disease later in life. However, the mechanisms linking the maternal obesogenic environment to adverse short- and long-term outcomes remain poorly understood. As compared with pregnant women with normal BMI, women entering pregnancy obese have more pronounced insulin resistance, higher circulating plasma insulin, leptin, IGF-1, lipids and possibly proinflammatory cytokines and lower plasma adiponectin. Importantly, the changes in maternal levels of nutrients, growth factors and hormones in maternal obesity modulate placental function. For example, high insulin, leptin, IGF-1 and low adiponectin in obese pregnant women activate mTOR signaling in the placenta, promoting protein synthesis, mitochondrial function and nutrient transport. These changes are believed to increase fetal nutrient supply and contribute to fetal overgrowth and/or adiposity in offspring, which increases the risk to develop disease later in life. However, the majority of obese women give birth to normal weight infants and these pregnancies are also associated with activation of inflammatory signaling pathways, oxidative stress, decreased oxidative phosphorylation and lipid accumulation in the placenta. Recent bioinformatics approaches have expanded our understanding of how maternal obesity affects the placenta; however, the link between changes in placental function and adverse outcomes in obese women giving birth to normal sized infants is unclear. Interventions that specifically target placental function, such as activation of placental adiponectin receptors, may prevent the transmission of metabolic disease from obese women to the next generation.


Assuntos
Obesidade Materna/metabolismo , Placenta/metabolismo , Complicações na Gravidez/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Adiposidade , Animais , Feminino , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Obesidade Materna/genética , Obesidade Materna/psicologia , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/fisiopatologia
6.
Proc Natl Acad Sci U S A ; 114(40): E8402-E8410, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28923943

RESUMO

The yeast prions [PSI+] and [URE3] are folded in-register parallel ß-sheet amyloids of Sup35p and Ure2p, respectively. In a screen for antiprion systems curing [PSI+] without protein overproduction, we detected Siw14p as an antiprion element. An array of genetic tests confirmed that many variants of [PSI+] arising in the absence of Siw14p are cured by restoring normal levels of the protein. Siw14p is a pyrophosphatase specifically cleaving the ß phosphate from 5-diphosphoinositol pentakisphosphate (5PP-IP5), suggesting that increased levels of this or some other inositol polyphosphate favors [PSI+] propagation. In support of this notion, we found that nearly all variants of [PSI+] isolated in a WT strain were lost upon loss of ARG82, which encodes inositol polyphosphate multikinase. Inactivation of the Arg82p kinase by D131A and K133A mutations (preserving Arg82p's nonkinase transcription regulation functions) resulted the loss of its ability to support [PSI+] propagation. The loss of [PSI+] in arg82Δ is independent of Hsp104's antiprion activity. [PSI+] variants requiring Arg82p could propagate in ipk1Δ (IP5 kinase), kcs1Δ (IP6 5-kinase), vip1Δ (IP6 1-kinase), ddp1Δ (inositol pyrophosphatase), or kcs1Δ vip1Δ mutants but not in ipk1Δ kcs1Δ or ddp1Δ kcs1Δ double mutants. Thus, nearly all [PSI+] prion variants require inositol poly-/pyrophosphates for their propagation, and at least IP6 or 5PP-IP4 can support [PSI+] propagation.


Assuntos
Inositol/metabolismo , Polifosfatos/metabolismo , Príons/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Príons/genética , Biossíntese de Proteínas , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/genética
7.
Trop Anim Health Prod ; 52(6): 3457-3466, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32939704

RESUMO

Sheep production in desert environments during summer is challenging due to heat stress which reduces feed intake, growth, and fertility. Despite warm conditions, some ewes are able to maintain a normal performance suggesting the existence of genetic bases underlying heat tolerance. Our objective was to discover and validate genetic markers associated with thermo-tolerance in pregnant ewes exposed to warm environmental conditions. Using a well-defined model laboratory of heat stress in sheep, pregnant Columbia-Rambouillet crossbred ewes (n = 100) were examined. Following acclimation to the laboratory at thermo-neutral conditions, heat stress was induced in ewes by increasing the temperature-humidity index in a control environmental chamber during mid-gestation. Feed intake, water consumption, and rectal temperature were recorded daily and used to establish the heat stress tolerance index (HSTI) for each ewe. Rectal temperature was a predictor (P < 0.05) of feed intake, and the regression coefficient was used to classify the HSTI. In a subset of 24 ewes, a genome-wide association study (GWAS) was performed using the Illumina OvineSNP50 BeadChip. Single-marker analysis detected 3 intragenic SNPs associated with HSTI (P value = 10-5). Bayesian multi-marker approach discovered 26 chromosomal regions across the genome which accounted for 9.8% of the variation associated with HSTI. In an independent sheep population (n = 42), the three discovered SNPs were validated as molecular markers associated with thermo-tolerance phenotypic traits. These SNPs were located within the genes F13A1, PAM, and PRELID2. In conclusion, three SNPs appear to be novel molecular markers associated with heat stress tolerance in pregnant ewes providing new knowledge about genetic foundations of thermo-tolerance.


Assuntos
Marcadores Genéticos/fisiologia , Resposta ao Choque Térmico/genética , Polimorfismo de Nucleotídeo Único/fisiologia , Carneiro Doméstico/fisiologia , Animais , Arizona , Feminino , Estudo de Associação Genômica Ampla/veterinária , Temperatura Alta , Gravidez , Carneiro Doméstico/genética , Termotolerância/genética
8.
J Physiol ; 597(24): 5835-5858, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31665811

RESUMO

KEY POINTS: Previous studies in fetuses with intrauterine growth restriction (IUGR) have shown that adrenergic dysregulation was associated with low insulin concentrations and greater insulin sensitivity. Although whole-body glucose clearance is normal, 1-month-old lambs with IUGR at birth have higher rates of hindlimb glucose uptake, which may compensate for myocyte deficiencies in glucose oxidation. Impaired glucose-stimulated insulin secretion in IUGR lambs is due to lower intra-islet insulin availability and not from glucose sensing. We investigated adrenergic receptor (ADR) ß2 desensitization by administering oral ADRß modifiers for the first month after birth to activate ADRß2 and antagonize ADRß1/3. In IUGR lambs ADRß2 activation increased whole-body glucose utilization rates and insulin sensitivity but had no effect on isolated islet or myocyte deficiencies. IUGR establishes risk for developing diabetes. In IUGR lambs we identified disparities in key aspects of glucose-stimulated insulin secretion and insulin-stimulated glucose oxidation, providing new insights into potential mechanisms for this risk. ABSTRACT: Placental insufficiency causes intrauterine growth restriction (IUGR) and disturbances in glucose homeostasis with associated ß adrenergic receptor (ADRß) desensitization. Our objectives were to measure insulin-sensitive glucose metabolism in neonatal lambs with IUGR and to determine whether daily treatment with ADRß2 agonist and ADRß1/ß3 antagonists for 1 month normalizes their glucose metabolism. Growth, glucose-stimulated insulin secretion (GSIS) and glucose utilization rates (GURs) were measured in control lambs, IUGR lambs and IUGR lambs treated with adrenergic receptor modifiers: clenbuterol atenolol and SR59230A (IUGR-AR). In IUGR lambs, islet insulin content and GSIS were less than in controls; however, insulin sensitivity and whole-body GUR were not different from controls. Of importance, ADRß2 stimulation with ß1/ß3 inhibition increases both insulin sensitivity and whole-body glucose utilization in IUGR lambs. In IUGR and IUGR-AR lambs, hindlimb GURs were greater but fractional glucose oxidation rates and ex vivo skeletal muscle glucose oxidation rates were lower than controls. Glucose transporter 4 (GLUT4) was lower in IUGR and IUGR-AR skeletal muscle than in controls but GLUT1 was greater in IUGR-AR. ADRß2, insulin receptor, glycogen content and citrate synthase activity were similar among groups. In IUGR and IUGR-AR lambs heart rates were greater, which was independent of cardiac ADRß1 activation. We conclude that targeted ADRß2 stimulation improved whole-body insulin sensitivity but minimally affected defects in GSIS and skeletal muscle glucose oxidation. We show that risk factors for developing diabetes are independent of postnatal catch-up growth in IUGR lambs as early as 1 month of age and are inherent to the islets and myocytes.


Assuntos
Retardo do Crescimento Fetal/tratamento farmacológico , Resistência à Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Receptores Adrenérgicos beta 2/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/administração & dosagem , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Agonistas de Receptores Adrenérgicos beta 2/uso terapêutico , Antagonistas de Receptores Adrenérgicos beta 2/administração & dosagem , Antagonistas de Receptores Adrenérgicos beta 2/farmacocinética , Antagonistas de Receptores Adrenérgicos beta 2/uso terapêutico , Animais , Atenolol/administração & dosagem , Atenolol/farmacologia , Atenolol/uso terapêutico , Células Cultivadas , Clembuterol/administração & dosagem , Clembuterol/farmacologia , Clembuterol/uso terapêutico , Feminino , Retardo do Crescimento Fetal/metabolismo , Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Músculo Esquelético/metabolismo , Ovinos
9.
Xenotransplantation ; 25(6): e12432, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30052287

RESUMO

BACKGROUND: There is currently a shortage of human donor pancreata which limits the broad application of islet transplantation as a treatment for type 1 diabetes. Porcine islets have demonstrated potential as an alternative source, but a study evaluating islets from different donor ages under unified protocols has yet to be conducted. METHODS: Neonatal porcine islets (NPI; 1-3 days), juvenile porcine islets (JPI; 18-21 days), and adult porcine islets (API; 2+ years) were compared in vitro, including assessments of oxygen consumption rate, membrane integrity determined by FDA/PI staining, ß-cell proliferation, dynamic glucose-stimulated insulin secretion, and RNA sequencing. RESULTS: Oxygen consumption rate normalized to DNA was not significantly different between ages. Membrane integrity was age dependent, and API had the highest percentage of intact cells. API also had the highest glucose-stimulated insulin secretion response during a dynamic insulin secretion assay and had 50-fold higher total insulin content compared to NPI and JPI. NPI and JPI had similar glucose responsiveness, ß-cell percentage, and ß-cell proliferation rate. Transcriptome analysis was consistent with physiological assessments. API transcriptomes were enriched for cellular metabolic and insulin secretory pathways, while NPI exhibited higher expression of genes associated with proliferation. CONCLUSIONS: The oxygen demand, membrane integrity, ß-cell function and proliferation, and transcriptomes of islets from API, JPI, and NPI provide a comprehensive physiological comparison for future studies. These assessments will inform the optimal application of each age of porcine islet to expand the availability of islet transplantation.


Assuntos
Sobrevivência de Enxerto/imunologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Consumo de Oxigênio/fisiologia , Animais , Animais Recém-Nascidos , Diabetes Mellitus Experimental/terapia , Rejeição de Enxerto/imunologia , Células Secretoras de Insulina/imunologia , Transplante das Ilhotas Pancreáticas/métodos , Pâncreas/imunologia , Pâncreas/metabolismo , Suínos , Transcriptoma/imunologia , Transplante Heterólogo/métodos
10.
J Physiol ; 595(11): 3331-3343, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28144955

RESUMO

KEY POINTS: Thyroid hormones are important regulators of growth and maturation before birth, although the extent to which their actions are mediated by insulin and the development of pancreatic beta cell mass is unknown. Hypothyroidism in fetal sheep induced by removal of the thyroid gland caused asymmetric organ growth, increased pancreatic beta cell mass and proliferation, and was associated with increased circulating concentrations of insulin and leptin. In isolated fetal sheep islets studied in vitro, thyroid hormones inhibited beta cell proliferation in a dose-dependent manner, while high concentrations of insulin and leptin stimulated proliferation. The developing pancreatic beta cell is therefore sensitive to thyroid hormone, insulin and leptin before birth, with possible consequences for pancreatic function in fetal and later life. The findings of this study highlight the importance of thyroid hormones during pregnancy for normal development of the fetal pancreas. ABSTRACT: Development of pancreatic beta cell mass before birth is essential for normal growth of the fetus and for long-term control of carbohydrate metabolism in postnatal life. Thyroid hormones are also important regulators of fetal growth, and the present study tested the hypotheses that thyroid hormones promote beta cell proliferation in the fetal ovine pancreatic islets, and that growth retardation in hypothyroid fetal sheep is associated with reductions in pancreatic beta cell mass and circulating insulin concentration in utero. Organ growth and pancreatic islet cell proliferation and mass were examined in sheep fetuses following removal of the thyroid gland in utero. The effects of triiodothyronine (T3 ), insulin and leptin on beta cell proliferation rates were determined in isolated fetal ovine pancreatic islets in vitro. Hypothyroidism in the sheep fetus resulted in an asymmetric pattern of organ growth, pancreatic beta cell hyperplasia, and elevated plasma insulin and leptin concentrations. In pancreatic islets isolated from intact fetal sheep, beta cell proliferation in vitro was reduced by T3 in a dose-dependent manner and increased by insulin at high concentrations only. Leptin induced a bimodal response whereby beta cell proliferation was suppressed at the lowest, and increased at the highest, concentrations. Therefore, proliferation of beta cells isolated from the ovine fetal pancreas is sensitive to physiological concentrations of T3 , insulin and leptin. Alterations in these hormones may be responsible for the increased beta cell proliferation and mass observed in the hypothyroid sheep fetus and may have consequences for pancreatic function in later life.


Assuntos
Proliferação de Células , Doenças Fetais/fisiopatologia , Hiperinsulinismo/fisiopatologia , Hipotireoidismo/fisiopatologia , Células Secretoras de Insulina/fisiologia , Animais , Células Cultivadas , Feminino , Doenças Fetais/sangue , Hiperinsulinismo/sangue , Hiperinsulinismo/etiologia , Hipotireoidismo/sangue , Hipotireoidismo/complicações , Insulina/sangue , Células Secretoras de Insulina/efeitos dos fármacos , Leptina/sangue , Gravidez , Ovinos , Tri-Iodotironina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA