Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Islets ; 6(5-6): e1017226, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25833251

RESUMEN

Emerging evidence suggests that activin with its associated receptors, second messengers, and antagonists would be excellent targets for therapeutic drug development in the treatment of diabetes. We undertook the current study to investigate the ability to extrapolate findings from rodent studies to human islets in which data thus far has been scarce. We tested the hypothesis that human islets synthesize activin and that activin participates in the regulation of islet ß-cells. Human islets from 33 separate isolations were categorized based on functional status, culture status and diabetic status. Statistical comparisons were made by ANOVA with Tukey post-hoc adjustment for multiple comparisons. Experiments investigating activin utilized qPCR, FACS cell sorting, immunofluorescent antibody staining, functionality assays, viability assays and protein secretion assays. We have defined the transcript expression patterns of activin and the TGFß superfamily in human islets. We found INHBA (the gene encoding activin A) to be the most highly expressed of the superfamily in normal, cultured islets. We elucidated a link between the islet microenvironment and activin A. We found differential ligand expression based on diabetic, culture and functional status. Further, this is also the first report that links direct effects of activin A with the ability to restore glucose-stimulated insulin secretion in human islets from type 2 diabetic donors thereby establishing the relevance of targeting activin for therapeutic drug development.


Asunto(s)
Activinas/fisiología , Diabetes Mellitus Tipo 2/fisiopatología , Células Secretoras de Insulina/fisiología , Activinas/biosíntesis , Adulto , Anciano , Supervivencia Celular/fisiología , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Citometría de Flujo , Expresión Génica/fisiología , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/fisiología , Humanos , Insulina/biosíntesis , Células Secretoras de Insulina/metabolismo , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/fisiología , Adulto Joven
2.
Endocrinology ; 154(6): 2025-33, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23533219

RESUMEN

The development of insulin resistance is tightly linked to fatty liver disease and is considered a major health concern worldwide, although their mechanistic relationship remains controversial. Activin has emerging roles in nutrient homeostasis, but its metabolic effects on hepatocytes remain unknown. In this study, we investigated the effects of increased endogenous activin bioactivity on hepatic nutrient homeostasis by creating mice with inactivating mutations that deplete the circulating activin antagonists follistatin-like-3 (FSTL3) or the follistatin 315 isoform (FST315; FST288-only mice). We investigated liver histology and lipid content, hepatic insulin sensitivity, and metabolic gene expression including the HepG2 cell and primary hepatocyte response to activin treatment. Both FSTL3-knockout and FST288-only mice had extensive hepatic steatosis and elevated hepatic triglyceride content. Unexpectedly, insulin signaling, as assessed by phospho-Akt (a.k.a. protein kinase B), was enhanced in both mouse models. Pretreatment of HepG2 cells with activin A increased their response to subsequent insulin challenge. Gene expression analysis suggests that increased lipid uptake, enhanced de novo lipid synthesis, decreased lipolysis, and/or enhanced glucose uptake contribute to increased hepatic triglyceride content in these models. However, activin treatment recapitulated only some of these gene changes, suggesting that increased activin bioactivity may be only partially responsible for this phenotype. Nevertheless, our results indicate that activin enhances hepatocyte insulin response, which ultimately leads to hepatic steatosis despite the increased insulin sensitivity. Thus, regulation of activin bioactivity is critical for maintaining normal liver lipid homeostasis and response to insulin, whereas activin agonists may be useful for increasing liver insulin sensitivity.


Asunto(s)
Activinas/farmacocinética , Hígado Graso/metabolismo , Resistencia a la Insulina , Hígado/metabolismo , Activinas/farmacología , Animales , Disponibilidad Biológica , Células Cultivadas , Hígado Graso/patología , Proteínas Relacionadas con la Folistatina , Expresión Génica/efectos de los fármacos , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Immunoblotting , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Fosfolipasas A2 Calcio-Independiente/genética , Fosfolipasas A2 Calcio-Independiente/metabolismo , Proteínas/genética , Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Triglicéridos/metabolismo
3.
Am J Physiol Endocrinol Metab ; 303(5): E587-96, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22739106

RESUMEN

Based on the phenotype of the activin-like kinase-7 (ALK7)-null mouse, activins A and B have been proposed to play distinct roles in regulating pancreatic islet function and glucose homeostasis, with activin A acting to enhance islet function and insulin release while activin B antagonizes these actions. We therefore hypothesized that islets from activin B-null (BBKO) mice would have enhanced glucose-stimulated insulin secretion. In addition, we hypothesized that this enhanced islet function would translate into increased whole body glucose tolerance. We tested these hypotheses by analyzing glucose homeostasis, insulin secretion, and islet function in BBKO mice. No differences were observed in fasting glucose or insulin levels, glucose tolerance, or insulin sensitivity compared with weight-matched young or older males. Similarly, there were no significant differences in insulin secretion comparing islets from WT or BBKO males at either age. However, BBKO islets were more sensitive to activin A, myostatin (MSTN), and follistatin (FST) treatments, so that activin A and FST inhibited and MSTN enhanced glucose stimulated insulin secretion. While mean islet area and the distribution of islet areas were not different between the genotypes, islet mass, islet number, and the proportion of α-cells/islet were significantly reduced in BBKO islets. These results indicate that activin B does not antagonize activin A to influence whole body glucose homeostasis or ß-cell function but does influence islet mass and proportion of α-cells/islet. Therefore, loss of activin B signaling alone does not account for the ALK7-null phenotype, but activin B may have important roles in modulating islet mass, islet number, and the cellular composition of islets.


Asunto(s)
Activinas/metabolismo , Envejecimiento/metabolismo , Homeostasis , Subunidades beta de Inhibinas/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Islotes Pancreáticos/citología , Activinas/genética , Envejecimiento/sangre , Animales , Glucemia/análisis , Recuento de Células , Folistatina/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Subunidades beta de Inhibinas/genética , Insulina/sangre , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Miostatina/metabolismo , Tamaño de los Órganos , Técnicas de Cultivo de Tejidos
4.
Islets ; 3(6): 367-75, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21964310

RESUMEN

Members of the TGFß superfamily, including activins and TGFß, modulate glucose-stimulated insulin secretion (GSIS) in vitro using rat islets while genetic manipulations that reduce TGFß superfamily signaling in vivo in mice produced hypoplastic islets and/or hyperglycemia. Moreover, deletion of Fstl3, an antagonist of activin and myostatin, resulted in enlarged islets and ß-cell hyperplasia. These studies suggest that endogenous TGFß superfamily ligands regulate ß-cell generation and/or function. To test this hypothesis, we examined endogenous TGFß ligand synthesis and action in isolated rat and mouse islets. We found that activin A, TGFß1, and myostatin treatment enhanced rat islet GSIS but none of the ligands tested enhanced GSIS in mouse islets. However, follistatin inhibited GSIS, consistent with a role for endogenous TGFß superfamily ligands in regulating insulin secretion. Endogenous expression of TGFß superfamily members was different in rat and mouse islets with myostatin being highly expressed in mouse islets and not detectable in rats. These results indicate that TGFß superfamily members directly regulate islet function in a species-specific manner while the ligands produced by islets differ between mice and rats. The lack of in vitro actions of ligands on mouse islets may be mechanical or result from species-specific actions of these ligands.


Asunto(s)
Activinas/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Miostatina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Activinas/genética , Animales , Proteínas Relacionadas con la Folistatina , Inmunohistoquímica , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miostatina/genética , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/química , ARN Mensajero/genética , Ratas , Ratas Endogámicas Lew , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/genética
5.
Obesity (Silver Spring) ; 19(10): 1940-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21546932

RESUMEN

Transforming growth factor-ß superfamily ligands, including activin and myostatin, modulate body composition, islet function, and glucose homeostasis. Their bioactivity is controlled by the antagonists follistatin (FST) and FST like-3 (FSTL3). The hypothesis tested was that FST and FSTL3 have distinct roles in regulating body composition, glucose homeostasis, and islet function through regulation of activin and myostatin bioactivity. Three genetic mutant mouse lines were created. FSTL3 knockout (FSTL3 KO), a mouse line producing only the FST288 isoform (FST288-only) and a double mutant (2xM) in which the lines were crossed. FST288-only males were lighter that wild-type (WT) littermates while FSTL3 KO and 2xM males had reduced perigonadal fat pad weights. However, only 2xM mice had increased whole body fat mass and decreased lean mass by quantitative nuclear magnetic resonance (qNMR). Fasting glucose levels in FSTL3 WT and KO mice were lower than FST mice in younger animals but were higher in older mice. Serum insulin and pancreatic insulin content in 2xM mice was significantly elevated over other genotypes. Nevertheless, 2xM mice were relatively insulin resistant and glucose intolerant compared to FST288-only and WT mice. Fractional islet area and proportion of ß-cells/islet were increased in FSTL3 KO and 2xM, but not FST288-only mice. Despite their larger size, islets from FSTL3 KO and 2xM mice were not functionally enhanced compared to WT mice. These results demonstrate that body composition and glucose homeostasis are differentially regulated by FST and FSTL3 and that their combined loss is associated with increased fat mass and insulin resistance despite elevated insulin production.


Asunto(s)
Tejido Adiposo/metabolismo , Glucemia/metabolismo , Proteínas Relacionadas con la Folistatina/metabolismo , Folistatina/metabolismo , Resistencia a la Insulina/fisiología , Mutación , Obesidad/metabolismo , Animales , Composición Corporal/genética , Composición Corporal/fisiología , Compartimentos de Líquidos Corporales/metabolismo , Peso Corporal/fisiología , Ayuno , Folistatina/genética , Proteínas Relacionadas con la Folistatina/genética , Genotipo , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Resistencia a la Insulina/genética , Islotes Pancreáticos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Páncreas/metabolismo , Isoformas de Proteínas
6.
Endocrinology ; 152(2): 697-706, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21106872

RESUMEN

Follistatin (FST) is an antagonist of activin and related TGFß superfamily members that has important reproductive actions as well as critical regulatory functions in other tissues and systems. FST is produced as three protein isoforms that differ in their biochemical properties and in their localization within the body. We created FST288-only mice that only express the short FST288 isoform and previously reported that females are subfertile, but have an excess of primordial follicles on postnatal day (PND) 8.5 that undergo accelerated demise in adults. We have now examined germ cell nest breakdown and primordial follicle formation in the critical PND 0.5-8.5 period to test the hypothesis that the excess primordial follicles derive from increased proliferation and decreased apoptosis during germ cell nest breakdown. Using double immunofluorescence microscopy we found that there is virtually no germ cell proliferation after birth in wild-type or FST288-only females. However, the entire process of germ cell nest breakdown was extended in time (through at least PND 8.5) and apoptosis was significantly reduced in FST288-only females. In addition, FST288-only females are born with more germ cells within the nests. Thus, the excess primordial follicles in FST288-only mice derive from a greater number of germ cells at birth as well as a reduced rate of apoptosis during nest breakdown. These results also demonstrate that FST is critical for normal regulation of germ cell nest breakdown and that loss of the FST303 and/or FST315 isoforms leads to excess primordial follicles with accelerated demise, resulting in premature cessation of ovarian function.


Asunto(s)
Células Germinativas/citología , Células Germinativas/metabolismo , Folículo Ovárico/citología , Folículo Ovárico/metabolismo , Isoformas de Proteínas/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Femenino , Técnica del Anticuerpo Fluorescente , Folistatina , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Isoformas de Proteínas/genética
7.
Endocrinology ; 151(3): 1310-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20032047

RESUMEN

Follistatin (FST) is a natural antagonist of activin and related TGFbeta superfamily ligands that exists as three protein isoforms differing in length at the C terminus. The longest FST315 isoform is found in the circulation, whereas the shortest FST288 isoform is typically found in or on cells and tissues, and the intermediate FST303 isoform is found in gonads. We recently demonstrated that the FST isoforms have distinct biological actions in vitro that, taken together with the differential distribution, suggests they may also have different roles in vivo. To explore the specific role of individual FST isoforms, we created a single-isoform FST288-only mouse. In contrast to the neonatal death of FST global knockout mice, FST288-only mice survive to adulthood. Although they appear normal, FST288-only mice have fertility defects including reduced litter size and frequency. Follicles were counted in ovaries from 8.5- to 400-d-old females. Significantly fewer morphologically healthy antral follicles were found in 100- to 250-d FST288-only ovaries, but there were significantly more secondary, primary, and primordial follicles detected at d 8.5 in FST288-only ovaries. However, depletion of this primordial follicle pool is more rapid in FST288-only females resulting in a deficit by 250 d of age and early cessation of reproduction. Superovulated FST288-only females have fewer ovulated eggs and embryos. These results indicate that the FST isoforms have different activities in vivo, that the FST288-only isoform is sufficient for development, and that loss of FST303 and FST315 isoforms results in fertility defects that resemble activin hyperactivity and premature ovarian failure.


Asunto(s)
Desarrollo Embrionario , Fertilidad , Folistatina/metabolismo , Ovario/metabolismo , Reproducción , Animales , Femenino , Ratones , Isoformas de Proteínas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...