Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 333
Filter
1.
Endocrinology ; 162(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-33972988

ABSTRACT

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.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Eating/drug effects , Ghrelin/pharmacology , Growth Hormone/blood , Receptors, Somatotropin/genetics , Receptors, Somatotropin/physiology , Agouti-Related Protein/analysis , Animals , Arcuate Nucleus of Hypothalamus/chemistry , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptide Y/genetics , Paraventricular Hypothalamic Nucleus/chemistry , Proto-Oncogene Proteins c-fos/analysis , RNA, Messenger/analysis , Receptors, Ghrelin/genetics , Receptors, Somatotropin/deficiency , Signal Transduction/physiology
2.
Endocrinology ; 162(6)2021 06 01.
Article in English | MEDLINE | ID: mdl-33693673

ABSTRACT

In healthy conditions, prepubertal growth follows an individual specific growth channel. Growth hormone (GH) is undoubtedly the major regulator of growth. However, the homeostatic regulation to maintain the individual specific growth channel during growth is unclear. We recently hypothesized a body weight sensing homeostatic regulation of body weight during adulthood, the gravitostat. We now investigated if sensing of body weight also contributes to the strict homeostatic regulation to maintain the individual specific growth channel during prepubertal growth. To evaluate the effect of increased artificial loading on prepubertal growth, we implanted heavy (20% of body weight) or light (2% of the body weight) capsules into the abdomen of 26-day-old male rats. The body growth, as determined by change in biological body weight and growth of the long bones and the axial skeleton, was reduced in rats bearing a heavy load compared with light load. Removal of the increased load resulted in a catch-up growth and a normalization of body weight. Loading decreased hypothalamic growth hormone releasing hormone mRNA, liver insulin-like growth factor (IGF)-1 mRNA, and serum IGF-1, suggesting that the reduced body growth was caused by a negative feedback regulation on the somatotropic axis and this notion was supported by the fact that increased loading did not reduce body growth in GH-treated rats. Based on these data, we propose the gravitostat hypothesis for the regulation of prepubertal growth. This states that there is a homeostatic regulation to maintain the individual specific growth channel via body weight sensing, regulating the somatotropic axis and explaining catch-up growth.


Subject(s)
Body Weight/physiology , Growth Hormone/pharmacology , Growth and Development/drug effects , Animals , Body Weight/drug effects , Growth Hormone/metabolism , Growth Hormone-Releasing Hormone/metabolism , Homeostasis/drug effects , Locomotion/physiology , Male , Rats , Rats, Sprague-Dawley , Receptors, Somatotropin/drug effects , Receptors, Somatotropin/metabolism , Receptors, Somatotropin/physiology , Sexual Maturation/drug effects , Signal Transduction/drug effects
3.
J Endocrinol ; 248(1): 31-44, 2021 01.
Article in English | MEDLINE | ID: mdl-33112796

ABSTRACT

A reduction in hepatocyte growth hormone (GH)-signaling promotes non-alcoholic fatty liver disease (NAFLD). However, debate remains as to the relative contribution of the direct effects of GH on hepatocyte function vs indirect effects, via alterations in insulin-like growth factor 1 (IGF1). To isolate the role of hepatocyte GH receptor (GHR) signaling, independent of changes in IGF1, mice with adult-onset, hepatocyte-specific GHR knockdown (aHepGHRkd) were treated with a vector expressing rat IGF1 targeted specifically to hepatocytes. Compared to GHR-intact mice, aHepGHRkd reduced circulating IGF1 and elevated GH. In male aHepGHRkd, the shift in IGF1/GH did not alter plasma glucose or non-esterified fatty acids (NEFA), but was associated with increased insulin, enhanced systemic lipid oxidation and reduced white adipose tissue (WAT) mass. Livers of male aHepGHRkd exhibited steatosis associated with increased de novo lipogenesis, hepatocyte ballooning and inflammation. In female aHepGHRkd, hepatic GHR protein levels were not detectable, but moderate levels of IGF1 were maintained, with minimal alterations in systemic metabolism and no evidence of steatosis. Reconstitution of hepatocyte IGF1 in male aHepGHRkd lowered GH and normalized insulin, whole body lipid utilization and WAT mass. However, IGF1 reconstitution did not reduce steatosis or eliminate liver injury. RNAseq analysis showed IGF1 reconstitution did not impact aHepGHRkd-induced changes in liver gene expression, despite changes in systemic metabolism. These results demonstrate the impact of aHepGHRkd is sexually dimorphic and the steatosis and liver injury observed in male aHepGHRkd mice is autonomous of IGF1, suggesting GH acts directly on the adult hepatocyte to control NAFLD progression.


Subject(s)
Fatty Liver/etiology , Growth Hormone/physiology , Hepatocytes/physiology , Insulin-Like Growth Factor I/physiology , Liver/metabolism , Animals , Female , Lipid Metabolism , Male , Mice , Receptors, Somatotropin/physiology , Sex Characteristics , Somatotrophs/metabolism
4.
Front Endocrinol (Lausanne) ; 11: 579909, 2020.
Article in English | MEDLINE | ID: mdl-33162937

ABSTRACT

Growth hormone (GH) signaling plays a key role in mediating growth, development, metabolism, and lifespan regulation. However, the mechanisms of longevity regulation at the cellular and molecular level are still not well-understood. An important area in the field of GH research is in the development of advanced transgenic systems for conditional expression of GH signaling in a cell type- or tissue-specific manner. There have been many recent studies conducted to examine the effects of tissue-specific GHR disruption. This review updates our previous discussions on this topic and summarizes recent data on the newly-made tissue-specific GHR-KO mice including intestinal epithelial cells, bone, hematopoietic stem cells, cardiac myocytes, and specific brain regions. The data from these new genetically-engineered mice have a significant impact on our understanding of the local GH signaling function.


Subject(s)
Growth Hormone/metabolism , Homeostasis , Receptors, Somatotropin/physiology , Animals , Longevity , Mice , Mice, Knockout , Organ Specificity
5.
Mol Cell Endocrinol ; 518: 110999, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32835785

ABSTRACT

In this review, I summarize historical and recent features of the classical pathways activated by growth hormone (GH) through the cell surface GH receptor (GHR). GHR is a cytokine receptor superfamily member that signals by activating the non-receptor tyrosine kinase, JAK2, and members of the Src family kinases. Activation of the GHR engages STATs, PI3K, and ERK pathways, among others, and details of these now-classical pathways are presented. Modulating elements, including the SOCS proteins, phosphatases, and regulated GHR metalloproteolysis, are discussed. In addition, a novel physical and functional interaction of GHR with IGF-1R is summarized and discussed in terms of its mechanisms, consequences, and physiological and therapeutic implications.


Subject(s)
Receptors, Somatotropin/metabolism , Animals , Growth Hormone/metabolism , Growth Hormone/physiology , Human Growth Hormone/metabolism , Human Growth Hormone/physiology , Humans , Receptors, Somatotropin/physiology , Signal Transduction/physiology
6.
Mol Metab ; 36: 100978, 2020 06.
Article in English | MEDLINE | ID: mdl-32277923

ABSTRACT

OBJECTIVE: The liver is a central target organ of growth hormone (GH), which stimulates the synthesis of insulin-like growth factor 1 (IGF1) and affects multiple biochemical pathways. A systematic multi-omics analysis of GH effects in the liver has not been performed. GH receptor (GHR) deficiency is a unique model for studying the consequences of lacking GH action. In this study, we used molecular profiling techniques to capture a broad spectrum of these effects in the liver of a clinically relevant large animal model for Laron syndrome. METHODS: We performed holistic proteome and targeted metabolome analyses of liver samples from 6-month-old GHR-deficient (GHR-KO) pigs and GHR-expressing controls (four males, four females per group). RESULTS: GHR deficiency resulted in an increased abundance of enzymes involved in amino acid degradation, in the urea cycle, and in the tricarboxylic acid cycle. A decreased ratio of long-chain acylcarnitines to free carnitine suggested reduced activity of carnitine palmitoyltransferase 1A and thus reduced mitochondrial import of fatty acids for beta-oxidation. Increased levels of short-chain acylcarnitines in the liver and in the circulation of GHR-KO pigs may result from impaired beta-oxidation of short-chain fatty acids or from increased degradation of specific amino acids. The concentration of mono-unsaturated glycerophosphocholines was significantly increased in the liver of GHR-KO pigs without morphological signs of steatosis, although the abundances of several proteins functionally linked to non-alcoholic fatty liver disease (fetuin B, retinol binding protein 4, several mitochondrial proteins) were increased. Moreover, GHR-deficient liver samples revealed distinct changes in the methionine and glutathione metabolic pathways, in particular, a significantly increased level of glycine N-methyltransferase and increased levels of total and free glutathione. Several proteins revealed a sex-related abundance difference in the control group but not in the GHR-KO group. CONCLUSIONS: Our integrated proteomics/targeted metabolomics study of GHR-deficient and control liver samples from a clinically relevant large animal model identified a spectrum of biological pathways that are significantly altered in the absence of GH action. Moreover, new insights into the role of GH in the sex-related specification of liver functions were provided.


Subject(s)
Growth Hormone/metabolism , Liver/physiology , Receptors, Somatotropin/metabolism , Animals , Female , Gene Knockout Techniques/methods , Growth Hormone/physiology , Laron Syndrome , Male , Metabolomics/methods , Models, Animal , Non-alcoholic Fatty Liver Disease/metabolism , Protein Binding , Protein Transport , Proteomics/methods , Receptors, Somatotropin/genetics , Receptors, Somatotropin/physiology , Signal Transduction , Swine
7.
Arch. endocrinol. metab. (Online) ; 63(6): 557-567, Nov.-Dec. 2019. tab, graf
Article in English | LILACS | ID: biblio-1055021

ABSTRACT

ABSTRACT In order to provide new insights into the various activities of GH in specific tissues, recent advances have allowed for the generation of tissue-specific GHR knockout mice. To date, 21 distinct tissue-specific mouse lines have been created and reported in 28 publications. Targeted tissues include liver, muscle, fat, brain, bone, heart, intestine, macrophage, pancreatic beta cells, hematopoietic stem cells, and multi-tissue "global". In this review, we provide a brief history and description of the 21 tissue-specific GHR knockout mouse lines. Arch Endocrinol Metab. 2019;63(6):557-67


Subject(s)
Animals , Rats , Receptors, Somatotropin/physiology , Growth Hormone/physiology , Signal Transduction , Mice, Knockout , Models, Animal
8.
Growth Horm IGF Res ; 46-47: 5-15, 2019.
Article in English | MEDLINE | ID: mdl-31078722

ABSTRACT

OBJECTIVE: Growth hormone (GH) has been reported to enhance the intestinal barrier; as such, recombinant GH has been administered for several intestinal diseases. However, excess GH action has been implicated in increasing the risk of intestinal dysfunction. The goal of this study was to examine the direct effects of GH on the small and large intestines to clarify the role GH plays in intestinal function through the use of a mouse model. DESIGN: An intestinal epithelial-specific GH receptor (GHR) knockout (IntGHRKO) mouse line was generated using Cre-lox with the villin promoter driving Cre expression. The generated mice were characterized with respect to growth and intestinal phenotypes. RESULTS: IntGHRKO mice showed no significant changes in body length, weight, or composition compared to floxed controls. Male IntGHRKO mice had significantly shorter large intestines at 4 and 12 months of age. Intestinal barrier function was assessed by measuring the expression of tight junction related genes, as well as levels of serum endotoxin and fecal albumin. Results showed sex differences as males had an increase in occludin levels but normal serum endotoxin and fecal albumin; while, females had changes in fecal albumin levels with normal occludin and serum endotoxin. Evaluation of glucose tolerance and fat absorption also showed sex differences as females were glucose intolerant, while males had impaired fat absorption. Histopathology revealed a trend towards decreased villus height in males, which could explain the sex difference in glucose homeostasis. CONCLUSIONS: Overall, the data demonstrate that disruption of GH on the intestinal epithelial cells modestly affects the intestinal gross anatomy, morphology, and function in a sex-specific manner.


Subject(s)
Glucose/metabolism , Homeostasis , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Intestinal Mucosa/metabolism , Receptors, Somatotropin/physiology , Animals , Body Weight , Female , Gene Knockout Techniques , Integrases/metabolism , Male , Mice , Mice, Knockout
9.
J Neuroendocrinol ; 31(4): e12692, 2019 04.
Article in English | MEDLINE | ID: mdl-30712287

ABSTRACT

In patients with growth hormone (GH) deficiency (GHD), low doses of recombinant human GH (rhGH) have a similar or better long-term clinical effect than higher doses. Pharmacogenetic studies suggest that GH receptor (GHR) polymorphism only influences some metabolic parameters. Nonetheless, there is no clear scientific evidence proving the effects of lower rhGH dose regimens on metabolic parameters. The aim of our prospective study was to evaluate the effects of GHR polymorphism in adult GHD patients treated with low rhGH dose during short- (6 and 12 months) and long-term (5 years) follow-up. Sixty-nine GHD adult patients were studied, before and during treatment with rhGH, using a standardised low-dose protocol calculated on the basis of body weight (0.01-0.03 mg kg-1  week-1 ) and monitored by an insulin-like growth factor (IGF)-I plasma assay, as well as anthropometric and metabolic parameters. The GHR genotype (flfl, fld3 or d3d3) was determined from the peripheral blood. d3-GHR carriers showed a more effective short- and long-term response to low rhGH dose with respect to low-density lipoprotein reduction, body composition and blood pressure (homozygous patients only); d3-GHR homozygosity is related to a significant IGF-I increase during short-term follow-up. Regression analysis demonstrated that rhGH dose, age at diagnosis and GHR genotype are the major determinants of IGF-I increase at 6 and 12 months of replacement therapy. The d3d3-GHR genotype may influence some metabolic effects during the short- and long-term follow-up of low rhGH dose and could be an independent determinant of the increase of IGF- I during short-term follow-up.


Subject(s)
Human Growth Hormone/administration & dosage , Human Growth Hormone/deficiency , Polymorphism, Genetic/genetics , Receptors, Somatotropin/genetics , Receptors, Somatotropin/physiology , Recombinant Proteins/administration & dosage , Adult , Aged , Female , Genotype , Hormone Replacement Therapy , Humans , Insulin-Like Growth Factor I/analysis , Male , Middle Aged , Polymorphism, Genetic/physiology , Prospective Studies
10.
Arch Endocrinol Metab ; 63(6): 557-567, 2019.
Article in English | MEDLINE | ID: mdl-31939480

ABSTRACT

In order to provide new insights into the various activities of GH in specific tissues, recent advances have allowed for the generation of tissue-specific GHR knockout mice. To date, 21 distinct tissue-specific mouse lines have been created and reported in 28 publications. Targeted tissues include liver, muscle, fat, brain, bone, heart, intestine, macrophage, pancreatic beta cells, hematopoietic stem cells, and multi-tissue "global". In this review, we provide a brief history and description of the 21 tissue-specific GHR knockout mouse lines. Arch Endocrinol Metab. 2019;63(6):557-67.


Subject(s)
Growth Hormone/physiology , Receptors, Somatotropin/physiology , Animals , Mice , Mice, Knockout , Models, Animal , Signal Transduction
11.
Aging (Albany NY) ; 10(9): 2243-2251, 2018 09 16.
Article in English | MEDLINE | ID: mdl-30222593

ABSTRACT

It is widely accepted that caloric restriction (CR) extends lifespan and suppresses various pathophysiological changes. CR suppresses growth hormone/insulin-like growth factor signaling and mechanistic target of rapamycin complex 1 activity, activates sirtuin and enhances mitochondrial redox regulation, but the exact mechanisms are still under debate. In this review, we discuss the mechanisms of CR using evidence from studies of animals that were genetically modified according to recent advances in molecular and genetic technologies, from the viewpoint of the adaptive response hypothesis proposed by Holliday (1989). We then explain the beneficial actions of CR, classified according to whether they operate under feeding or fasting conditions.


Subject(s)
Aging , Caloric Restriction , Longevity , Adipose Tissue, White/metabolism , Animals , Animals, Genetically Modified , Mice , NF-E2-Related Factor 2/physiology , Neuropeptide Y/physiology , Receptors, Somatotropin/physiology , Sirtuins/physiology , TOR Serine-Threonine Kinases/physiology
12.
Proc Natl Acad Sci U S A ; 115(7): E1495-E1503, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29378959

ABSTRACT

It is well documented that inhibition of mTORC1 (defined by Raptor), a complex of mechanistic target of rapamycin (mTOR), extends life span, but less is known about the mechanisms by which mTORC2 (defined by Rictor) impacts longevity. Here, rapamycin (an inhibitor of mTOR) was used in GHR-KO (growth hormone receptor knockout) mice, which have suppressed mTORC1 and up-regulated mTORC2 signaling, to determine the effect of concurrently decreased mTORC1 and mTORC2 signaling on life span. We found that rapamycin extended life span in control normal (N) mice, whereas it had the opposite effect in GHR-KO mice. In the rapamycin-treated GHR-KO mice, mTORC2 signaling was reduced without further inhibition of mTORC1 in the liver, muscle, and s.c. fat. Glucose and lipid homeostasis were impaired, and old GHR-KO mice treated with rapamycin lost functional immune cells and had increased inflammation. In GHR-KO MEF cells, knockdown of Rictor, but not Raptor, decreased mTORC2 signaling. We conclude that drastic reduction of mTORC2 plays important roles in impaired longevity in GHR-KO mice via disruption of whole-body homeostasis.


Subject(s)
Immunosuppressive Agents/pharmacology , Longevity/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Receptors, Somatotropin/physiology , Sirolimus/pharmacology , Animals , Cytoplasm/drug effects , Cytoplasm/metabolism , Female , Insulin Resistance , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction
13.
Growth Horm IGF Res ; 38: 3-7, 2018 02.
Article in English | MEDLINE | ID: mdl-29198419

ABSTRACT

To elucidate whether a specific tissue is responsible for the beneficial health and longevity phenotype seen in growth hormone (GH) receptor (R) knockout (GHRKO) mice, the GHR gene was disrupted specifically in insulin sensitive tissues; namely, liver, adipose, and muscle. Furthermore, to investigate if the health- and life-span effects seen in the germline GHRKO mice were replicated when GH action was ablated after puberty; young, adult onset GHRKO mice were produced and characterized. In this review, we summarized the main findings derived from these mouse lines.


Subject(s)
Insulin Resistance , Longevity , Receptors, Somatotropin/physiology , Animals , Mice , Mice, Knockout , Signal Transduction
14.
Sheng Li Xue Bao ; 69(5): 541-556, 2017 Oct 25.
Article in English | MEDLINE | ID: mdl-29063103

ABSTRACT

Growth hormone (GH), as a vital hormone, has to experience a series of processes to fulfill its function including secretion, entering the circulation to reach target tissues (pre-receptor process), binding on the GH receptor (GHR) and triggering signaling inside cells (post-GHR process). Insulin can directly or indirectly influence part of these processes. GH secretion from pituitary somatotropes is regulated by GH-releasing hormone (GHRH) and somatostatin (SS) from hypothalamus. Insulin may exert positive or negative effects on the neurons expressing GHRH and SS and somatotropes under healthy and pathological conditions including obesity and diabetes. Glucose and lipid levels in circulation and dietary habits may influence the effect of insulin on GH secretion. Insulin may also affect GHR sensitivity and the level of insulin-like growth factor 1 (IGF-1), thus influence the level of GH. The GH signaling is also important for GH to play its role. GH signaling involves GHR/JAK2/STATs, GHR/JAK2/SHC/MAPK and GH/insulin receptor substrate (IRS)/PI3K/Akt pathways. These pathways may be shared by insulin, which is the basis for the interaction between insulin and GH, and insulin may attenuate or facilitate the GH signal by influencing molecules in the pathways. Many factors are related to the effect of insulin, among them the most important ones are duration of action and amount of insulin. The tendency of insulin-reduced GH signaling becomes obvious with increased dose and acting time of insulin. The participation of suppressor of cytokine signaling (SOCS), the interaction between JAK2 and IRS, and GHR sensitivity should also be considered when discovering GH signal. The involvement of SS in response to insulin is not clear yet. The details of how GH secretion, level and signaling change in response to time and dose of insulin treatment warrant further studies.


Subject(s)
Growth Hormone/metabolism , Insulin/pharmacology , Signal Transduction/drug effects , Animals , Growth Hormone/analysis , Growth Hormone/physiology , Humans , Janus Kinase 2/physiology , Receptors, Somatotropin/physiology , Signal Transduction/physiology
15.
J Gerontol A Biol Sci Med Sci ; 72(3): 329-337, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27208894

ABSTRACT

Growth hormone receptor knockout (GHR-KO) mice are long lived with improved health span, making this an excellent model system for understanding biochemical mechanisms important to cognitive reserve. The purpose of the present study was to elucidate differences in cognition and glutamatergic dynamics between aged (20- to 24-month-old) GHR-KO and littermate controls. Glutamate plays a critical role in hippocampal learning and memory and is implicated in several neurodegenerative disorders, including Alzheimer's disease. Spatial learning and memory were assessed using the Morris water maze (MWM), whereas independent dentate gyrus (DG), CA3, and CA1 basal glutamate, release, and uptake measurements were conducted in isoflurane anesthetized mice utilizing an enzyme-based microelectrode array (MEA) coupled with constant potential amperometry. These MEAs have high temporal and low spatial resolution while causing minimal damage to the surrounding parenchyma. Littermate controls performed worse on the memory portion of the MWM behavioral task and had elevated DG, CA3, and CA1 basal glutamate and stimulus-evoked release compared with age-matched GHR-KO mice. CA3 basal glutamate negatively correlated with MWM performance. These results support glutamatergic regulation in learning and memory and may have implications for therapeutic targets to delay the onset of, or reduce cognitive decline, in Alzheimer's disease.


Subject(s)
Aging/physiology , Cognition/physiology , Glutamic Acid/physiology , Receptors, Somatotropin/physiology , Signal Transduction/physiology , Animals , Female , Memory , Mice , Mice, Knockout , Models, Animal , Spatial Learning
16.
J Gerontol A Biol Sci Med Sci ; 72(8): 1054-1061, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-27688483

ABSTRACT

Disruption of the growth hormone (GH) signaling pathway promotes insulin sensitivity and is associated with both delayed aging and extended longevity. Two kinds of long-lived mice-Ames dwarfs (df/df) and GH receptor gene-disrupted knockouts (GHRKO) are characterized by a suppressed GH axis with a significant reduction of body size and decreased plasma insulin-like growth factor-1 (IGF-1) and insulin levels. Ames dwarf mice are deficient in GH, prolactin, and thyrotropin, whereas GHRKOs are GH resistant and are dwarf with decreased circulating IGF-1 and increased GH. Crossing Ames dwarfs and GHRKOs produced a new mouse line (df/KO), lacking both GH and GH receptor. These mice are characterized by improved glucose tolerance and increased adiponectin level, which could imply that these mice should be also characterized by additional life-span extension when comparing with GHRKOs and Ames dwarfs. Importantly, our longevity experiments showed that df/KO mice maintain extended longevity when comparing with N control mice; however, they do not live longer than GHRKO and Ames df/df mice. These important findings indicate that silencing GH signal is important to extend the life span; however, further decrease of body size in mice with already inhibited GH signal does not extend the life span regardless of improved some health-span markers.


Subject(s)
Aging/physiology , Growth Hormone/physiology , Receptors, Somatotropin/physiology , Animals , Insulin/metabolism , Insulin Resistance/physiology , Insulin-Like Growth Factor I/metabolism , Life Expectancy , Mice , Mice, Knockout , Models, Animal , Signal Transduction/physiology
17.
Prog Mol Biol Transl Sci ; 138: 27-39, 2016.
Article in English | MEDLINE | ID: mdl-26940385

ABSTRACT

Growth hormone (GH)-induced signaling results in numerous effects in multiple tissues throughout the body. Elucidation of several of these effects has come from studies observing the various phenotypes of the GH receptor (GHR) gene-disrupted (GHR-/-) mouse. These mice are dwarf and obese with increased insulin sensitivity, are resistant to cancer and diabetes, and have an extended lifespan. While these mice have proven to be a valuable tool for understanding the pleiotropic effects of GH, we and others have generated novel tissue-specific GHR gene-disrupted mouse lines that are now helping to further dissect the actions of GH on specific cells/tissues. In this chapter, we summarize the various phenotypes observed in these mice.


Subject(s)
Receptors, Somatotropin/physiology , Adipose Tissue/metabolism , Animals , Islets of Langerhans/metabolism , Liver/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Muscles/metabolism , Phenotype , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism
18.
Aging (Albany NY) ; 8(3): 539-46, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26959761

ABSTRACT

Caloric restriction and genetic disruption of growth hormone signaling have been shown to counteract aging in mice. The effects of these interventions on aging are examined through age-dependent survival or through the increase in age-dependent mortality rates on a logarithmic scale fitted to the Gompertz model. However, these methods have limitations that impede a fully comprehensive disclosure of these effects. Here we examine the effects of these interventions on murine aging through the increase in age-dependent mortality rates on a linear scale without fitting them to a model like the Gompertz model. Whereas these interventions negligibly and non-consistently affected the aging rates when examined through the age-dependent mortality rates on a logarithmic scale, they caused the aging rates to increase at higher ages and to higher levels when examined through the age-dependent mortality rates on a linear scale. These results add to the debate whether these interventions postpone or slow aging and to the understanding of the mechanisms by which they affect aging. Since different methods yield different results, it is worthwhile to compare their results in future research to obtain further insights into the effects of dietary, genetic, and other interventions on the aging of mice and other species.


Subject(s)
Aging , Caloric Restriction , Receptors, Somatotropin/physiology , Survival Analysis , Animals , Female , Gene Targeting , Male , Mice, 129 Strain
19.
J Dairy Res ; 82(3): 287-92, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26119533

ABSTRACT

In addition to the main components of the somatotrophic axis (GH/GHR/IGF-I/IGF-IR), great importance in the control of growth and development is also attached to the Janus kinase 2 (JAK2) pathway. Induced by the GH/GHR complex, JAK2 activates signal transducer and activator of transcription 5 (STAT5), and in consequence, may be involved in the regulation of expression of insulin-like growth factor I (IGF-I) in the mammary gland. Silent mutation (rs110298451) has been identified within exon 20 using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). A total of 904 individuals of four dairy or dual-purpose breeds (Polish Holstein-Friesian, Montbeliarde, Simmental and Jersey) were genotyped. A genotypic imbalance in the populations was observed. In the case of dual-purpose breeds (Montbeliarde and Simmental), the frequencies of both alleles were almost equal. In contrary, the JAK2G allele was predominant in the Polish Holstein-Friesian breed while JAK2A allele in Jersey. A pronounced relationship between JAK2/RsaI polymorphism and milk production traits was found where, irrespective of breed and lactation order, the GG genotype was significantly associated with higher milk, protein and fat yields, as compared to the AA genotype. Heterozygous individuals were generally characterised by intermediate values of the analysed milk traits. It can be argued that the JAK2 gene polymorphism is a potential marker for milk production traits. However, due to the fact that rs110298451 SNP does not directly affect amino acid sequence, other association studies involving missense mutation should also be performed.


Subject(s)
Cattle/genetics , Genetic Markers/genetics , Janus Kinase 2/genetics , Lactation/genetics , Quantitative Trait Loci/genetics , Animals , Breeding , Cattle/physiology , Fats/analysis , Female , Gene Expression , Genotype , Growth Hormone/physiology , Insulin-Like Growth Factor I/genetics , Mammary Glands, Animal/metabolism , Milk/chemistry , Milk Proteins/analysis , Polymerase Chain Reaction/veterinary , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide/genetics , Quantitative Trait, Heritable , Receptors, Somatotropin/physiology , STAT5 Transcription Factor/metabolism
20.
Theriogenology ; 82(1): 27-35, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24725418

ABSTRACT

This study aimed to demonstrate the expression of growth hormone receptor (GH-R) mRNA and protein in goat ovarian follicles in order to investigate the effects of GH on the survival and development of preantral follicles. The ovaries were processed for the isolation of follicles to study GH-R mRNA expression or to localization of GH-R by immunohistochemical analysis. Pieces of ovarian cortex were cultured for 7 days in minimum essential medium(+) (MEM(+)) in the presence or absence of GH at different concentrations (1, 10, 50, 100, and 200 ng/mL). High expression levels of GH-R mRNA were observed in granulosa/theca cells from large antral follicles. However, preantral follicles do not express mRNA for GH-R. Immunohistochemistry demonstrated that the GH-R protein was expressed in the oocytes/granulosa cells of antral follicles, but any protein expression was observed in preantral follicles. The highest (P < 0.05) rate of normal follicles and intermediate follicles was observed after 7 days in MEM(+) plus 10 ng/mL GH (70%). In conclusion, GH-R mRNA and protein are expressed in caprine antral follicles, but not in preantral follicles. Moreover, GH maintains the survival of goat preantral follicles and promotes the development of primordial follicles.


Subject(s)
Goats/physiology , Growth Hormone/pharmacology , Ovarian Follicle/growth & development , Receptors, Somatotropin/metabolism , Animals , Cell Culture Techniques , Female , Goats/genetics , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Ovarian Follicle/drug effects , RNA, Messenger/metabolism , Receptors, Somatotropin/physiology , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...