Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Endocrinology ; 162(7)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33972988

RESUMEN

Ghrelin stimulates both GH secretion and food intake. The orexigenic action of ghrelin is mainly mediated by neurons that coexpress agouti-related protein (AgRP) and neuropeptide Y (NPY) in the arcuate nucleus of the hypothalamus (ARH). GH also stimulates food intake and, importantly, ARHAgRP/NPY neurons express GH receptor (GHR). Thus, ghrelin-induced GH secretion may contribute to the orexigenic effect of ghrelin. Here, we investigated the response to ghrelin in male mice carrying GHR ablation specifically in neurons (brain GHR knockout [KO] mice) or exclusively in ARHAgRP/NPY neurons (AgRP GHR KO mice). Although brain GHR KO mice showed normal ghrelin-induced increase in plasma GH levels, these mutants lacked the expected orexigenic response to ghrelin. Additionally, brain GHR KO mice displayed reduced hypothalamic levels of Npy and Ghsr mRNA and did not elicit ghrelin-induced c-Fos expression in the ARH. Furthermore, brain GHR KO mice exhibited a prominent reduction in AgRP fiber density in the ARH and paraventricular nucleus of the hypothalamus (PVH). In contrast, AgRP GHR KO mice showed no changes in the hypothalamic Npy and Ghsr mRNAs and conserved ghrelin-induced food intake and c-Fos expression in the ARH. AgRP GHR KO mice displayed a reduced AgRP fiber density (~16%) in the PVH, but this reduction was less than that observed in brain GHR KO mice (~61%). Our findings indicate that GHR signaling in the brain is required for the orexigenic effect of ghrelin, independently of GH action on ARHAgRP/NPY neurons.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ghrelina/farmacología , Hormona del Crecimiento/sangre , Receptores de Somatotropina/genética , Receptores de Somatotropina/fisiología , Proteína Relacionada con Agouti/análisis , Animales , Núcleo Arqueado del Hipotálamo/química , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptido Y/genética , Núcleo Hipotalámico Paraventricular/química , Proteínas Proto-Oncogénicas c-fos/análisis , ARN Mensajero/análisis , Receptores de Ghrelina/genética , Receptores de Somatotropina/deficiencia , Transducción de Señal/fisiología
2.
Am J Physiol Endocrinol Metab ; 320(6): E1158-E1172, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33938235

RESUMEN

In the endocrine pancreas, growth hormone (GH) is known to promote pancreatic islet growth and insulin secretion. In this study, we show that GH receptor (GHR) loss in the germline and in adulthood impacts islet mass in general but more profoundly in male mice. GHR knockout (GHRKO) mice have enhanced insulin sensitivity and low circulating insulin. We show that the total cross-sectional area of isolated islets (estimated islet mass) was reduced by 72% in male but by only 29% in female GHRKO mice compared with wild-type controls. Also, islets from GHRKO mice secreted ∼50% less glucose-stimulated insulin compared with size-matched islets from wild-type mice. We next used mice with a floxed Ghr gene to knock down the GHR in adult mice at 6 mo of age (6mGHRKO) and examined the impact on glucose and islet metabolism. By 12 mo of age, female 6mGHRKO mice had increased body fat and reduced islet mass but had no change in glucose tolerance or insulin sensitivity. However, male 6mGHRKO mice had nearly twice as much body fat, substantially reduced islet mass, and enhanced insulin sensitivity, but no change in glucose tolerance. Despite large losses in islet mass, glucose-stimulated insulin secretion from isolated islets was not significantly different between male 6mGHRKO and controls, whereas isolated islets from female 6mGHRKO mice showed increased glucose-stimulated insulin release. Our findings demonstrate the importance of GH to islet mass throughout life and that unique sex-specific adaptations to the loss of GH signaling allow mice to maintain normal glucose metabolism.NEW & NOTEWORTHY Growth hormone (GH) is important for more than just growth. GH helps to maintain pancreatic islet mass and insulin secretion throughout life. Sex-specific adaptations to the loss of GH signaling allow mice to maintain normal glucose regulation despite losing islet mass.


Asunto(s)
Células Germinativas/metabolismo , Hormona del Crecimiento/deficiencia , Islotes Pancreáticos/crecimiento & desarrollo , Islotes Pancreáticos/fisiología , Receptores de Somatotropina/genética , Factores de Edad , Animales , Proliferación Celular/genética , Femenino , Células Germinativas/fisiología , Hormona del Crecimiento/genética , Hormona del Crecimiento/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos/genética , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/metabolismo , Caracteres Sexuales , Transducción de Señal/genética
3.
Aging Cell ; 19(5): e13123, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32110850

RESUMEN

Growth hormone receptor knockout (GHRKO) mice are remarkably long-lived and have improved glucose homeostasis along with altered energy metabolism which manifests through decreased respiratory quotient (RQ) and increased oxygen consumption (VO2 ). Short-term exposure of these animals to increased environmental temperature (eT) at 30°C can normalize their VO2 and RQ. We hypothesized that increased heat loss in the diminutive GHRKO mice housed at 23°C and the consequent metabolic adjustments to meet the increased energy demand for thermogenesis may promote extension of longevity, and preventing these adjustments by chronic exposure to increased eT will reduce or eliminate their longevity advantage. To test these hypotheses, GHRKO mice were housed at increased eT (30°C) since weaning. Here, we report that contrasting with the effects of short-term exposure of adult GHRKO mice to 30°C, transferring juvenile GHRKO mice to chronic housing at 30°C did not normalize the examined parameters of energy metabolism and glucose homeostasis. Moreover, despite decreased expression levels of thermogenic genes in brown adipose tissue (BAT) and elevated core body temperature, the lifespan of male GHRKO mice was not reduced, while the lifespan of female GHRKO mice was increased, along with improved glucose homeostasis. The results indicate that GHRKO mice have intrinsic features that help maintain their delayed, healthy aging, and extended longevity at both 23°C and 30°C.


Asunto(s)
Longevidad , Receptores de Somatotropina/metabolismo , Temperatura , Animales , Metabolismo Energético , Femenino , Glucosa/metabolismo , Homeostasis , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Receptores de Somatotropina/deficiencia , Destete
4.
J Mol Endocrinol ; 63(2): 123-138, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31357177

RESUMEN

It has been hypothesized that transcriptional changes associated with lower mTORC1 activity in mice with reduced levels of growth hormone and insulin-like growth factor 1 are responsible for the longer healthy lifespan of these mutant mice. Cell lines and tissues from these mice show alterations in the levels of many proteins that cannot be explained by corresponding changes in mRNAs. Such post-transcriptional modulation may be the result of preferential mRNA translation by the cap-independent translation of mRNA bearing the N6-methyl-adenosine (m6A) modification. The long-lived endocrine mutants - Snell dwarf, growth hormone receptor deletion and pregnancy-associated plasma protein-A knockout - all show increases in the N6-adenosine-methyltransferases (METTL3/14) that catalyze 6-methylation of adenosine (m6A) in the 5' UTR region of select mRNAs. In addition, these mice have elevated levels of YTH domain-containing protein 1 (YTHDF1), which recognizes m6A and promotes translation by a cap-independent mechanism. Consistently, multiple proteins that can be translated by the cap-independent mechanism are found to increase in these mice, including DNA repair and mitochondrial stress response proteins, without changes in corresponding mRNA levels. Lastly, a drug that augments cap-independent translation by inhibition of cap-dependent pathways (4EGI-1) was found to elevate levels of the same set of proteins and able to render cells resistant to several forms of in vitro stress. Augmented translation by cap-independent pathways facilitated by m6A modifications may contribute to the stress resistance and increased healthy longevity of mice with diminished GH and IGF-1 signals.


Asunto(s)
Sistema Endocrino/metabolismo , Longevidad , Mutación/genética , Biosíntesis de Proteínas , Caperuzas de ARN/metabolismo , Regulación hacia Arriba , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Fibroblastos/metabolismo , Humanos , Hidrazonas/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones Noqueados , Ratones Mutantes , Proteolisis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/metabolismo , Tiazoles/farmacología , Regulación hacia Arriba/efectos de los fármacos
5.
Mol Metab ; 11: 113-128, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29678421

RESUMEN

OBJECTIVE: Laron syndrome (LS) is a rare, autosomal recessive disorder in humans caused by loss-of-function mutations of the growth hormone receptor (GHR) gene. To establish a large animal model for LS, pigs with GHR knockout (KO) mutations were generated and characterized. METHODS: CRISPR/Cas9 technology was applied to mutate exon 3 of the GHR gene in porcine zygotes. Two heterozygous founder sows with a 1-bp or 7-bp insertion in GHR exon 3 were obtained, and their heterozygous F1 offspring were intercrossed to produce GHR-KO, heterozygous GHR mutant, and wild-type pigs. Since the latter two groups were not significantly different in any parameter investigated, they were pooled as the GHR expressing control group. The characterization program included body and organ growth, body composition, endocrine and clinical-chemical parameters, as well as signaling studies in liver tissue. RESULTS: GHR-KO pigs lacked GHR and had markedly reduced serum insulin-like growth factor 1 (IGF1) levels and reduced IGF-binding protein 3 (IGFBP3) activity but increased IGFBP2 levels. Serum GH concentrations were significantly elevated compared with control pigs. GHR-KO pigs had a normal birth weight. Growth retardation became significant at the age of five weeks. At the age of six months, the body weight of GHR-KO pigs was reduced by 60% compared with controls. Most organ weights of GHR-KO pigs were reduced proportionally to body weight. However, the weights of liver, kidneys, and heart were disproportionately reduced, while the relative brain weight was almost doubled. GHR-KO pigs had a markedly increased percentage of total body fat relative to body weight and displayed transient juvenile hypoglycemia along with decreased serum triglyceride and cholesterol levels. Analysis of insulin receptor related signaling in the liver of adult fasted pigs revealed increased phosphorylation of IRS1 and PI3K. In agreement with the loss of GHR, phosphorylation of STAT5 was significantly reduced. In contrast, phosphorylation of JAK2 was significantly increased, possibly due to the increased serum leptin levels and increased hepatic leptin receptor expression and activation in GHR-KO pigs. In addition, increased mTOR phosphorylation was observed in GHR-KO liver samples, and phosphorylation studies of downstream substrates suggested the activation of mainly mTOR complex 2. CONCLUSION: GHR-KO pigs resemble the pathophysiology of LS and are an interesting model for mechanistic studies and treatment trials.


Asunto(s)
Síndrome de Laron/genética , Hígado/metabolismo , Receptores de Somatotropina/genética , Transducción de Señal , Adiposidad , Animales , Peso Corporal , Hormona del Crecimiento/sangre , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Janus Quinasa 2/metabolismo , Síndrome de Laron/fisiopatología , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Receptores de Somatotropina/deficiencia , Factor de Transcripción STAT5/metabolismo , Porcinos
6.
Growth Horm IGF Res ; 38: 53-56, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29306560

RESUMEN

Recombinant human insulin-like growth factor-1 (rhIGF-1) treatment studies of growth failure in absence of growth hormone (GH) signaling (GH insensitivity -GHI, Laron syndrome -LS, GH Receptor deficiency -GHRD) have taken place in many locations around the globe. Results from these trials are comparable, and slight differences reported can be attributed to specific circumstances at different research sites. rhIGF-I treatment studies of GHI in Ecuador included various trials performed on children belonging to the largest and only homogeneous cohort of subjects with this condition in the world. All trials were performed by the same team of investigators and, during study periods, subjects received similar nutritional, physical activity and medical advice. Combination of these inherent conditions most likely creates less sources of variability during the research process. Indeed, diagnosis, selection and inclusion of research subjects; methodology used; transport, storage and delivery of study drug; data collection, monitoring and auditing; data analysis, discussion of results, conclusion inferences and reporting, etc., were submitted to the same sources of error. For the above-mentioned reasons, we are hereby mainly covering conclusions derived from rhIGF-I treatment studies of Ecuadorian children whit GHRD due to homozygosity of a splice site mutation occurring at GHR gene, whose unaffected parents were both heterozygous for the same mutation. We also describe studies of rhIGF-I administration in adolescent and adult subjects with GHRD, from the same cohort and with the same genetic anomaly.


Asunto(s)
Trastornos del Crecimiento/tratamiento farmacológico , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Receptores de Somatotropina/deficiencia , Ecuador/epidemiología , Trastornos del Crecimiento/epidemiología , Humanos
7.
Aging Cell ; 16(1): 52-60, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27618784

RESUMEN

Studies of the mTOR pathway have prompted speculation that diminished mTOR complex-1 (mTORC1) function may be involved in controlling the aging process. Our previous studies have shown diminished mTORC1 activity in tissues of three long-lived mutant mice: Snell dwarf mice, growth hormone receptor gene disrupted mice (GHRKO), and in this article, mice deficient in the pregnancy-associated protein-A (PAPPA-KO). The ways in which lower mTOR signals slow aging and age-related diseases are, however, not well characterized. Here, we show that Snell, GHKRO, and PAPPA-KO mice express high levels of two proteins involved in DNA repair, O-6-methylguanine-DNA methyltransferase (MGMT) and N-myc downstream-regulated gene 1 (NDRG1). Furthermore, we report that lowering mTOR enhances MGMT and NDRG1 protein expression via post-transcriptional mechanisms. We show that the CCR4-NOT complex, a post-transcriptional regulator of gene expression, is downstream of the mTORC1 pathway and may be responsible for the upregulation of MGMT and NDRG1 in all three varieties of long-lived mice. Our data thus suggest a novel link between DNA repair and mTOR signaling via post-transcriptional regulation involving specific alteration in the CCR4-NOT complex, whose modulation could control multiple aspects of the aging process.


Asunto(s)
Proteínas de Ciclo Celular/genética , Daño del ADN , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Enanismo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Longevidad , Proteína Plasmática A Asociada al Embarazo/deficiencia , Receptores de Somatotropina/deficiencia , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Proteínas de Ciclo Celular/metabolismo , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Noqueados , Modelos Biológicos , Proteína Plasmática A Asociada al Embarazo/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR4/metabolismo , Receptores de Somatotropina/metabolismo , Sirolimus/farmacología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
8.
Cell Rep ; 14(7): 1571-1580, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26876170

RESUMEN

The hallmarks of age-related immune senescence are chronic inflammation, aberrant expansion of effector memory, and loss of naive T lymphocytes due in part to systemic activation of innate immune sensor NLRP3 inflammasome in myeloid lineage cells. The endogenous mechanisms that regulate inflammasome activation during aging are unknown. Here, we present evidence that growth hormone receptor (GH-R)-dependent downregulation of NLRP3 inflammasome in macrophages is linked to pro-longevity effects that maintain immune system homeostasis in aging. Deletion of GH-R prevented the macrophage-driven age-related activation of inflammasome in response to NLRP3 ligands and also increased the preservation of naive T cells, even in advanced age and with higher IFNγ secretion from effector cells. The mechanism of inflammasome inhibition is linked to autocrine somatotropic axis as ablation of IGF1R in macrophages lowered the NLRP3 inflammasome activation. Together, our findings show that functional somatotropic axis in macrophages controls inflammation, thus linking NLRP3-mediated innate immune signaling to health span and longevity.


Asunto(s)
Envejecimiento/genética , Proteínas Portadoras/genética , Inflamasomas/genética , Macrófagos/inmunología , Receptor IGF Tipo 1/genética , Receptores de Somatotropina/genética , Envejecimiento/inmunología , Animales , Comunicación Autocrina , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Proteínas Portadoras/inmunología , Regulación de la Expresión Génica , Homeostasis/inmunología , Inmunidad Innata , Memoria Inmunológica , Inflamasomas/inmunología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Longevidad/genética , Longevidad/inmunología , Macrófagos/citología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/inmunología , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/inmunología , Transducción de Señal , Bazo/citología , Bazo/inmunología , Linfocitos T/citología , Linfocitos T/inmunología
9.
Endocrine ; 52(2): 194-205, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26732039

RESUMEN

Previous studies have shown that GH and IGF-I may enhance tumorigenesis, metastasis, and cell proliferation in humans and animals. Evidence supporting this notion is derived from animal model studies, epidemiological studies, experience from patients with acromegaly, molecular therapeutic manipulation of GH and IGF-I actions, and individuals with GH receptor and congenital IGF-I deficiencies. Prior exposure to radiation therapy, aging, family history of cancer, and individual susceptibility may also contribute to increase this risk. Therefore, the use of GH replacement in patients with a history of cancer raises hypothetical safety concerns for patients, caregivers, and providers. Studies of GH therapy in GH-deficient adults with hypopituitarism and childhood cancer survivors have not convincingly demonstrated an increased cancer risk. Conversely, the risk of occurrence of a second neoplasm (SN) in childhood cancer survivors may be increased, with meningiomas being the most common tumor; however, this risk appears to decline over time. In light of these findings, if GH replacement is to be considered in patients with a previous history of cancer, we propose this consideration to be based on each individual circumstance and that such therapy should only be initiated at least 2 years after cancer remission is achieved with the understanding that in some patients (particularly those with childhood cancers), GH may potentially increase the risk of SNs. In addition, close surveillance should be undertaken working closely with the patient's oncologist. More long-term data are thus needed to determine if GH replacement in GH-deficient adults with a history of cancer is associated with the development of de novo tumors and tumor recurrence.


Asunto(s)
Hormona del Crecimiento , Terapia de Reemplazo de Hormonas , Hipopituitarismo/tratamiento farmacológico , Neoplasias/inducido químicamente , Acromegalia/inducido químicamente , Animales , Contraindicaciones , Hormona del Crecimiento/administración & dosificación , Hormona del Crecimiento/efectos adversos , Terapia de Reemplazo de Hormonas/efectos adversos , Humanos , Factor I del Crecimiento Similar a la Insulina/deficiencia , Receptores de Somatotropina/deficiencia
10.
J Endocrinol ; 226(1): 13-23, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25943560

RESUMEN

GH influences adipocyte differentiation, but both stimulatory and inhibitory effects have been described. Adipose tissue-derived mesenchymal stem cells (AT-MSCs) are multipotent and are able to differentiate into adipocytes, among other cells. Canonical Wnt/ß-catenin signaling activation impairs adipogenesis. The aim of the present study was to elucidate the role of GH on AT-MSC adipogenesis using cells isolated from male GH receptor knockout (GHRKO), bovine GH transgenic (bGH) mice, and wild-type littermate control (WT) mice. AT-MSCs from subcutaneous (sc), epididiymal (epi), and mesenteric (mes) AT depots were identified and isolated by flow cytometry (Pdgfrα+ Sca1+ Cd45- Ter119- cells). Their in vitro adipogenic differentiation capacity was determined by cell morphology and real-time RT-PCR. Using identical in vitro conditions, adipogenic differentiation of AT-MSCs was only achieved in the sc depot, and not in epi and mes depots. Notably, we observed an increased differentiation in cells isolated from sc-GHRKO and an impaired differentiation of sc-bGH cells as compared to sc-WT cells. Axin2, a marker of Wnt/ß-catenin activation, was increased in mature sc-bGH adipocytes, which suggests that activation of this pathway may be responsible for the decreased adipogenesis. Thus, the present study demonstrates that (i) adipose tissue in mice has a well-defined population of Pdgfrα+ Sca1+ MSCs; (ii) the differentiation capacity of AT-MSCs varies from depot to depot regardless of GH genotype; (iii) the lack of GH action increases adipogenesis in the sc depot; and iv) activation of the Wnt/ß-catenin pathway might mediate the GH effect on AT-MSCs. Taken together, the present results suggest that GH diminishes fat mass in part by altering adipogenesis of MSCs.


Asunto(s)
Adipogénesis/fisiología , Tejido Adiposo Blanco/citología , Hormona del Crecimiento/fisiología , Células Madre Mesenquimatosas/citología , Adipocitos/citología , Animales , Antígenos Ly/metabolismo , Bovinos , Diferenciación Celular , Hormona del Crecimiento/genética , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo
11.
J Gerontol A Biol Sci Med Sci ; 70(1): 44-52, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24550353

RESUMEN

Apoptosis is a process that affects life span and health. Mice with liver-specific disruption of the growth hormone receptor (GHR) gene (ie, Ghr gene) liver-specific growth hormone receptor knockout [LiGHRKO] mice), as opposed to mice with global deletion of the Ghr gene (GHRKO; Ghr-/-), are characterized by severe hepatic steatosis and lack of improved insulin sensitivity. We have previously shown that levels of proapoptotic factors are decreased in long-lived and insulin-sensitive GHRKO mice. In the current study, expression of specific apoptosis-related genes was assessed in brains, kidneys, and livers of male and female LiGHRKO and wild-type mice using real-time PCR. In the brain, expression of Caspase 3, Caspase 9, Smac/DIABLO, and p53 was decreased in females compared with males. Renal expression of Caspase 3 and Noxa also decreased in female mice. In the liver, no differences were seen between males and females. Also, no significant genotype effects were detected in the examined organs. Lack of significant genotype effect in kidneys contrasts with previous observations in GHRKO mice. Apparently, global GHR deletion induces beneficial changes in apoptotic factors, whereas liver-specific GHR disruption does not. Furthermore, sexual dimorphism may play an important role in regulating apoptosis during liver-specific suppression of the somatotrophic signaling.


Asunto(s)
Apoptosis/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Hígado/metabolismo , Longevidad/genética , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/genética , Animales , Proteínas Reguladoras de la Apoptosis , Encéfalo/metabolismo , Caspasas/genética , Femenino , Genes bcl-2 , Genes p53 , Riñón/metabolismo , Hígado/citología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Mitocondriales/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Caracteres Sexuales , Proteína X Asociada a bcl-2/genética
12.
Proteomics ; 13(3-4): 674-85, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23019135

RESUMEN

Growth hormone (GH) is a protein secreted by the anterior pituitary and circulates throughout the body to exert important actions on growth and metabolism. GH stimulates the secretion of insulin-like growth factor-I (IGF-I) that mediates some of the growth promoting actions of GH. The GH/IGF-I axis has recently been recognized as important in terms of longevity in organisms ranging from Caenorhabditis elegans to mice. For example, GH transgenic mice possess short lifespans while GH receptor null (GHR-/-) mice have extended longevity. Thus, the actions of GH (or IGF-I) or lack thereof impact the aging process. In this review, we summarize the proteomic analyses of plasma and white adipose tissue in these two mouse models of GH action, i.e. GH transgenic and GHR-/- mice. At the protein level, we wanted to establish novel plasma biomarkers of GH action as a function of age and to determine differences in adipose tissue depots. We have shown that these proteomic approaches have not only confirmed several known physiological actions of GH, but also resulted in novel protein biomarkers and targets that may be indicative of the aging process and/or new functions of GH. These results may generate new directions for GH and/or aging research.


Asunto(s)
Envejecimiento/metabolismo , Hormona del Crecimiento/fisiología , Proteoma/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Proteínas Sanguíneas/metabolismo , Técnicas de Inactivación de Genes , Humanos , Insulina/fisiología , Resistencia a la Insulina , Factor I del Crecimiento Similar a la Insulina/fisiología , Metabolismo de los Lípidos , Proteómica , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/genética
13.
Aging Cell ; 11(4): 668-74, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22577861

RESUMEN

Previous work has shown that primary skin-derived fibroblasts from long-lived pituitary dwarf mutants resist the lethal effects of many forms of oxidative and nonoxidative stress. We hypothesized that increased autophagy may protect fibroblasts of Pit-1(dw/dw) (Snell dwarf) mice from multiple forms of stress. We found that dwarf-derived fibroblasts had higher levels of autophagy, using LC3 and p62 as markers, in response to amino acid deprivation, hydrogen peroxide, and paraquat. Fibroblasts from dwarf mice also showed diminished phosphorylation of mTOR, S6K, and 4EBP1, consistent with the higher levels of autophagy in these cells after stress. Similar results were also observed in fibroblasts from mutant mice lacking growth hormone receptor (GHRKO mice) after amino acid withdrawal. Our results suggested that increased autophagy, regulated by TOR-dependent processes, may contribute to stress resistance in fibroblasts from long-lived mutant mice.


Asunto(s)
Autofagia/genética , Longevidad/genética , Longevidad/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Autofagia/efectos de los fármacos , Autofagia/fisiología , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Medios de Cultivo , Factores Eucarióticos de Iniciación , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Peróxido de Hidrógeno/toxicidad , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Ratones Mutantes , Estrés Oxidativo , Paraquat/toxicidad , Fosfoproteínas/metabolismo , Fosforilación , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/genética , Receptores de Somatotropina/fisiología , Proteínas Quinasas S6 Ribosómicas/metabolismo , Factor de Transcripción Pit-1/deficiencia , Factor de Transcripción Pit-1/genética , Factor de Transcripción Pit-1/fisiología
14.
Gerontology ; 58(4): 337-43, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22261798

RESUMEN

A recent report of virtually complete protection from diabetes and cancer in a population of people with hereditary dwarfism revived interest in elucidating the relationships between growth, adult body size, age-related disease and longevity. In many species, smaller individuals outlive those that are larger and a similar relationship was shown in studies of various human populations. Adult body size is strongly dependent on the actions of growth hormone (GH) and the absence of GH or GH receptor in mice leads to a remarkable extension of longevity. Many mechanisms that may account for, or contribute to, this association have been identified. It is suggested that modest modifications of the diet at different ages may extend human healthspan and lifespan by reducing levels of hormones that stimulate growth.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Tamaño Corporal/fisiología , Longevidad/fisiología , Tejido Adiposo/fisiopatología , Animales , Enanismo/patología , Enanismo/fisiopatología , Hormona del Crecimiento/deficiencia , Hormona del Crecimiento/fisiología , Humanos , Insulina/deficiencia , Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina/deficiencia , Factor I del Crecimiento Similar a la Insulina/fisiología , Ratones , Modelos Animales , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/fisiología , Transducción de Señal , Serina-Treonina Quinasas TOR/deficiencia , Serina-Treonina Quinasas TOR/fisiología
15.
Endocrinology ; 153(1): 273-82, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22109889

RESUMEN

Fasting results in the mobilization of adipose stores and the elevation of levels of free fatty acids (FFA). In humans, this process is driven by a release in GH. Little is known regarding the role of GH in modulating this process during early stages of fasting in the mouse. Confirmation of the role of GH in modulating FFA release in the fasting mouse is of particular importance given the frequent use of mouse models to study metabolic mechanisms. Here, we correlate the initial release of FFA throughout fasting in mice with pulsatile GH secretion. Observations illustrate the rapid release of FFA in response to food withdrawal. This does not correlate with a rise in GH secretion. Rather, we observed a striking loss in pulsatile secretion of GH throughout the first 6 h of fasting, suggesting that GH does not modulate the initial release of FFA in the mouse in response to fasting. This was confirmed in GH receptor knockout mice, in which we observed a robust fasting-induced rise in FFA. We further illustrate the dynamic relationship between the orexigenic and anorexigenic hormones ghrelin and leptin during fasting in the mouse. Our findings show an initial suppression of leptin and the eventual rise in circulating levels of acyl-ghrelin with fasting. However, altered acyl-ghrelin and leptin secretion occurs well after the rise in FFA and the suppression of GH secretion. Consequently, we conclude that although acyl-ghrelin and leptin may modulate the physiological response to drive food intake, these changes do not contribute to the initial loss of pulsatile GH secretion. Rather, it appears that the suppression of GH secretion in fasting may occur in response to an elevation in fasting levels of FFA or physiological stress. Observations highlight a divergent role for GH in modulating FFA release between man and mouse.


Asunto(s)
Ayuno/fisiología , Ácidos Grasos no Esterificados/metabolismo , Hormona del Crecimiento/fisiología , Animales , Corticosterona/sangre , Ayuno/sangre , Ácidos Grasos no Esterificados/sangre , Expresión Génica , Ghrelina/sangre , Hormona del Crecimiento/sangre , Hormona del Crecimiento/metabolismo , Humanos , Hipotálamo/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Hipófisis/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/genética , Transducción de Señal , Especificidad de la Especie , Factores de Tiempo
16.
Aging Cell ; 11(1): 73-81, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22040032

RESUMEN

Mice with targeted deletion of the growth hormone receptor (GHRKO mice) are growth hormone (GH) resistant, small, obese, hypoinsulinemic, highly insulin sensitive and remarkably long-lived. To elucidate the unexpected coexistence of adiposity with improved insulin sensitivity and extended longevity, we examined effects of surgical removal of visceral (epididymal and perinephric) fat on metabolic traits related to insulin signaling and longevity. Comparison of results obtained in GHRKO mice and in normal animals from the same strain revealed disparate effects of visceral fat removal (VFR) on insulin and glucose tolerance, adiponectin levels, accumulation of ectopic fat, phosphorylation of insulin signaling intermediates, body temperature, and respiratory quotient (RQ). Overall, VFR produced the expected improvements in insulin sensitivity and reduced body temperature and RQ in normal mice and had opposite effects in GHRKO mice. Some of the examined parameters were altered by VFR in opposite directions in GHRKO and normal mice, and others were affected in only one genotype or exhibited significant genotype × treatment interactions. Functional differences between visceral fat of GHRKO and normal mice were confirmed by measurements of adipokine secretion, lipolysis, and expression of genes related to fat metabolism. We conclude that in the absence of GH signaling, the secretory activity of visceral fat is profoundly altered and unexpectedly promotes enhanced insulin sensitivity. The apparent beneficial effects of visceral fat in GHRKO mice may also explain why reducing adiposity by calorie restriction fails to improve insulin signaling or further extend longevity in these animals.


Asunto(s)
Insulina/metabolismo , Grasa Intraabdominal/metabolismo , Metabolismo de los Lípidos/genética , Longevidad/genética , Receptores de Somatotropina/genética , Transducción de Señal/genética , Adiponectina/biosíntesis , Animales , Biomarcadores/metabolismo , Temperatura Corporal , Restricción Calórica , Eliminación de Gen , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Hormona del Crecimiento/metabolismo , Resistencia a la Insulina/genética , Grasa Intraabdominal/cirugía , Masculino , Ratones , Ratones Noqueados , Fosforilación , Receptores de Somatotropina/deficiencia
17.
Exp Biol Med (Maywood) ; 236(2): 156-68, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21321312

RESUMEN

The long-lived growth hormone (GH) receptor knockout (GHRKO; KO) mice are GH-resistant due to targeted disruption of the GH receptor (Ghr) gene. Apoptosis is a physiological process in which cells play an active role in their own death and is a normal component of the development and health of multicellular organisms. Aging is associated with the progressive loss of strength of skeletal and heart muscles. Calorie restriction (CR) is a well-known experimental model to delay aging and increase lifespan. The aim of the study was to examine the expression of the following apoptosis-related genes: caspase-3, caspase-9, caspase-8, bax, bcl-2, Smac/DIABLO, p53 and cytochrome c1 (cyc1) in the skeletal muscles and hearts of female normal and GHRKO mice, fed ad libitum or subjected to 40% CR for six months, starting at two months of age. Moreover, skeletal muscle caspase-3, caspase-9, caspase-8, bax, bcl-2, Smac/DIABLO, Apaf-1, bad, phospho-bad (pbad), phospho-p53 and cytochrome c (cyc) protein expression levels were assessed. Expression of caspase-3, caspase-9, bax and Smac/DIABLO genes and proteins was decreased in GHRKO's skeletal muscles. The Apaf-1 protein expression also was diminished in this tissue. In contrast, bcl-2 and pbad protein levels were increased in skeletal muscles in knockouts. No changes were demonstrated for the examined genes' expression in GHRKO's hearts except for the increased level of cyc1 mRNA. CR did not alter the expression of the examined genes and proteins in skeletal muscles of knockouts versus normal (N) mice. In heart homogenates, CR increased caspase-3 mRNA level as compared with ad libitum mice. Decreased expression of certain proapoptotic genes and/or proteins may constitute the potential mechanism of prolonged longevity in GHRKO mice, protecting these animals from aging; this potential beneficial mechanism is not affected by CR.


Asunto(s)
Apoptosis , Restricción Calórica , Corazón/fisiopatología , Músculo Esquelético/fisiopatología , Receptores de Somatotropina/deficiencia , Animales , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados
18.
Sci Transl Med ; 3(70): 70ra13, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21325617

RESUMEN

Mutations in growth signaling pathways extend life span, as well as protect against age-dependent DNA damage in yeast and decrease insulin resistance and cancer in mice. To test their effect in humans, we monitored for 22 years Ecuadorian individuals who carry mutations in the growth hormone receptor (GHR) gene that lead to severe GHR and IGF-1 (insulin-like growth factor-1) deficiencies. We combined this information with surveys to identify the cause and age of death for individuals in this community who died before this period. The individuals with GHR deficiency exhibited only one nonlethal malignancy and no cases of diabetes, in contrast to a prevalence of 17% for cancer and 5% for diabetes in control subjects. A possible explanation for the very low incidence of cancer was suggested by in vitro studies: Serum from subjects with GHR deficiency reduced DNA breaks but increased apoptosis in human mammary epithelial cells treated with hydrogen peroxide. Serum from GHR-deficient subjects also caused reduced expression of RAS, PKA (protein kinase A), and TOR (target of rapamycin) and up-regulation of SOD2 (superoxide dismutase 2) in treated cells, changes that promote cellular protection and life-span extension in model organisms. We also observed reduced insulin concentrations (1.4 µU/ml versus 4.4 µU/ml in unaffected relatives) and a very low HOMA-IR (homeostatic model assessment-insulin resistance) index (0.34 versus 0.96 in unaffected relatives) in individuals with GHR deficiency, indicating higher insulin sensitivity, which could explain the absence of diabetes in these subjects. These results provide evidence for a role of evolutionarily conserved pathways in the control of aging and disease burden in humans.


Asunto(s)
Envejecimiento/fisiología , Diabetes Mellitus/epidemiología , Neoplasias/epidemiología , Receptores de Somatotropina/deficiencia , Adulto , Apoptosis/efectos de los fármacos , Apoptosis/genética , Células Cultivadas , Estudios de Cohortes , Roturas del ADN/efectos de los fármacos , Diabetes Mellitus/sangre , Diabetes Mellitus/metabolismo , Femenino , Genotipo , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/metabolismo , Receptores de Somatotropina/genética , Transducción de Señal/genética , Adulto Joven
19.
Endocr Rev ; 32(3): 356-86, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21123740

RESUMEN

Disruption of the GH receptor (GHR) gene eliminates GH-induced intracellular signaling and, thus, its biological actions. Therefore, the GHR gene disrupted mouse (GHR-/-) has been and is a valuable tool for helping to define various parameters of GH physiology. Since its creation in 1995, this mouse strain has been used by our laboratory and others for numerous studies ranging from growth to aging. Some of the most notable discoveries are their extreme insulin sensitivity in the presence of obesity. Also, the animals have an extended lifespan, which has generated a large number of investigations into the roles of GH and IGF-I in the aging process. This review summarizes the many results derived from the GHR-/- mice. We have attempted to present the findings in the context of current knowledge regarding GH action and, where applicable, to discuss how these mice compare to GH insensitivity syndrome in humans.


Asunto(s)
Receptores de Somatotropina/fisiología , Factores de Edad , Animales , Femenino , Hormona del Crecimiento/fisiología , Humanos , Resistencia a la Insulina , Factor I del Crecimiento Similar a la Insulina/fisiología , Masculino , Ratones , Ratones Noqueados , Obesidad/fisiopatología , Fenotipo , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/genética , Transducción de Señal
20.
J Endocrinol Invest ; 34(11): 861-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22322534

RESUMEN

The GH receptor (GHR) plays a key role in the the function of the GH/IGF-I axis and is the major effector of human growth. A common polymorphic variant consisting of genomic exon 3 deletion or retention (d3-GHR and full-length GHR, respectively), described in 2000, has been linked with increased receptor activity due to enhanced signal transduction. Subsequent pharmacogenetic studies have addressed a possible role of GHR polymorphism on the response to recombinant human GH treatment first in short children and then in adults, many of them suggesting that growth response to GH may be influenced, at least in some aspects, by this polymorphism. Similar studies, performed in patients with acromegaly, assumed an influence of the d3- GHR variant in the relationship between GH and IGF-I levels. More recently, some studies have investigated the relation between GHR genotype and treatment with the GHR antagonist pegvisomant, suggesting a better clinical response to therapy related to d3-GHR genotype. This review provides a summary of the main pharmacogenetic studies performed on this current and still open topic.


Asunto(s)
Exones/genética , Eliminación de Gen , Hormona de Crecimiento Humana/genética , Factor I del Crecimiento Similar a la Insulina/fisiología , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/genética , Acromegalia/genética , Acromegalia/patología , Animales , Hormona de Crecimiento Humana/fisiología , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Polimorfismo Genético/genética , Receptores de Somatotropina/fisiología , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...