Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 455
Filter
1.
Gen Comp Endocrinol ; 355: 114560, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38806133

ABSTRACT

Growth hormone-releasing hormone (GHRH) has been widely shown to stimulate growth hormone (GH) production via binding to GHRH receptor GHRHR in various species of vertebrates, but information regarding the functional roles of GHRH and GHRHR in the protochordate amphioxus remains rather scarce. We showed here that two mature peptides, BjGHRH-1 and BjGHRH-2, encoded by BjGHRH precursor, and a single BjGHRHR protein were identified in the amphioxus Branchiostoma. japonicum. Like the distribution profiles of vertebrate GHRHs and GHRHRs, both the genes Bjghrh and Bjghrhr were widely expressed in the different tissues of amphioxus, including in the cerebral vesicle, Hatschek's pit, neural tube, gill, hepatic caecum, notochord, testis and ovary. Moreover, both BjGHRH-1 and BjGHRH-2 interacted with BjGHRHR, and triggered the cAMP/PKA signal pathway in a dose-dependent manner. Importantly, BjGHRH-1 and BjGHRH-2 were both able to activate the expression of GH-like gene in the cells of Hatschek's pit. These indicate that a functional vertebrate-like GHRH-GHRHR axis had already emerged in amphioxus, which is a seminal innovation making physiological divergence including reproduction, growth, metabolism, stress and osmoregulation possible during the early evolution of vertebrates.


Subject(s)
Growth Hormone-Releasing Hormone , Lancelets , Receptors, Neuropeptide , Receptors, Pituitary Hormone-Regulating Hormone , Animals , Lancelets/metabolism , Lancelets/genetics , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/genetics , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Hypothalamo-Hypophyseal System/metabolism
2.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33468654

ABSTRACT

Therapies for heart failure with preserved ejection fraction (HFpEF) are lacking. Growth hormone-releasing hormone agonists (GHRH-As) have salutary effects in ischemic and nonischemic heart failure animal models. Accordingly, we hypothesized that GHRH-A treatment ameliorates chronic kidney disease (CKD)-induced HFpEF in a large-animal model. Female Yorkshire pigs (n = 16) underwent 5/6 nephrectomy via renal artery embolization and 12 wk later were randomized to receive daily subcutaneous injections of GHRH-A (MR-409; n = 8; 30 µg/kg) or placebo (n = 8) for 4 to 6 wk. Renal and cardiac structure and function were serially assessed postembolization. Animals with 5/6 nephrectomy exhibited CKD (elevated blood urea nitrogen [BUN] and creatinine) and faithfully recapitulated the hemodynamic features of HFpEF. HFpEF was demonstrated at 12 wk by maintenance of ejection fraction associated with increased left ventricular mass, relative wall thickness, end-diastolic pressure (EDP), end-diastolic pressure/end-diastolic volume (EDP/EDV) ratio, and tau, the time constant of isovolumic diastolic relaxation. After 4 to 6 wk of treatment, the GHRH-A group exhibited normalization of EDP (P = 0.03), reduced EDP/EDV ratio (P = 0.018), and a reduction in myocardial pro-brain natriuretic peptide protein abundance. GHRH-A increased cardiomyocyte [Ca2+] transient amplitude (P = 0.009). Improvement of the diastolic function was also evidenced by increased abundance of titin isoforms and their ratio (P = 0.0022). GHRH-A exerted a beneficial effect on diastolic function in a CKD large-animal model as demonstrated by improving hemodynamic, structural, and molecular characteristics of HFpEF. These findings have important therapeutic implications for the HFpEF syndrome.


Subject(s)
Cardiotonic Agents/pharmacology , Growth Hormone-Releasing Hormone/agonists , Heart Failure/drug therapy , Renal Insufficiency, Chronic/drug therapy , Sermorelin/analogs & derivatives , Stroke Volume/physiology , Animals , Blood Urea Nitrogen , Calcium/metabolism , Connectin/genetics , Connectin/metabolism , Creatinine/blood , Disease Models, Animal , Female , Gene Expression Regulation , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Heart Failure/etiology , Heart Failure/genetics , Heart Failure/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/genetics , Nephrectomy/methods , Peptide Fragments/blood , Peptide Fragments/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Sermorelin/pharmacology , Swine
3.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34782465

ABSTRACT

Ischemic stroke can induce neurogenesis. However, most stroke-generated newborn neurons cannot survive. It has been shown that MR-409, a potent synthetic agonistic analog of growth hormone-releasing hormone (GHRH), can protect against some life-threatening pathological conditions by promoting cell proliferation and survival. The present study shows that long-term treatment with MR-409 (5 or 10 µg/mouse/d) by subcutaneous (s.c.) injection significantly reduces the mortality, ischemic insult, and hippocampal atrophy, and improves neurological functional recovery in mice operated on for transient middle cerebral artery occlusion (tMCAO). Besides, MR-409 can stimulate endogenous neurogenesis and improve the tMCAO-induced loss of neuroplasticity. MR-409 also enhances the proliferation and inhibits apoptosis of neural stem cells treated with oxygen and glucose deprivation-reperfusion. The neuroprotective effects of MR-409 are closely related to the activation of AKT/CREB and BDNF/TrkB pathways. In conclusion, the present study demonstrates that GHRH agonist MR-409 has remarkable neuroprotective effects through enhancing endogenous neurogenesis in cerebral ischemic mice.


Subject(s)
Growth Hormone-Releasing Hormone/agonists , Growth Hormone-Releasing Hormone/metabolism , Ischemic Stroke/metabolism , Nerve Regeneration/drug effects , Neurogenesis/drug effects , Animals , Apoptosis/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Growth Hormone-Releasing Hormone/genetics , Infarction, Middle Cerebral Artery/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neuronal Plasticity , Neuroprotective Agents , Protein-Tyrosine Kinases/metabolism , Recovery of Function/drug effects
4.
Int J Mol Sci ; 25(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38892024

ABSTRACT

Inflammation, demyelination, and axonal damage to the central nervous system (CNS) are the hallmarks of multiple sclerosis (MS) and its representative animal model, experimental autoimmune encephalomyelitis (EAE). There is scientific evidence for the involvement of growth hormone (GH) in autoimmune regulation. Previous data on the relationship between the GH/insulin like growth factor-1 (IGF-1) axis and MS/EAE are inconclusive; therefore, the aim of our study was to investigate the changes in the GH axis during acute monophasic EAE. The results show that the gene expression of Ghrh and Sst in the hypothalamus does not change, except for Npy and Agrp, while at the pituitary level the Gh, Ghrhr and Ghr genes are upregulated. Interestingly, the cell volume of somatotropic cells in the pituitary gland remains unchanged at the peak of the disease. We found elevated serum GH levels in association with low IGF-1 concentration and downregulated Ghr and Igf1r expression in the liver, indicating a condition resembling GH resistance. This is likely due to inadequate nutrient intake at the peak of the disease when inflammation in the CNS is greatest. Considering that GH secretion is finely regulated by numerous central and peripheral signals, the involvement of the GH/IGF-1 axis in MS/EAE should be thoroughly investigated for possible future therapeutic strategies, especially with a view to improving EAE disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Growth Hormone , Insulin-Like Growth Factor I , Animals , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Rats , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/genetics , Hypothalamus/metabolism , Hypothalamus/pathology , Pituitary Gland/metabolism , Pituitary Gland/pathology , Receptors, Somatotropin/metabolism , Receptors, Somatotropin/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Multiple Sclerosis/genetics , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/genetics , Liver/metabolism , Liver/pathology , Disease Models, Animal
5.
Exp Lung Res ; 49(1): 152-164, 2023.
Article in English | MEDLINE | ID: mdl-37584484

ABSTRACT

Purpose: Growth hormone-releasing hormone (GHRH) is a 44-amino acid peptide that regulates growth hormone (GH) secretion. We hypothesized that GHRH receptor (GHRH-R) in alveolar type 2 (AT2) cells could modulate pro-inflammatory and possibly subsequent pro-fibrotic effects of lipopolysaccharide (LPS) or cytokines, such that AT2 cells could participate in lung inflammation and fibrosis. Methods: We used human alveolar type 2 (iAT2) epithelial cells derived from induced pluripotent stem cells (iPSC) to investigate how GHRH-R modulates gene and protein expression. We tested iAT2 cells' gene expression in response to LPS or cytokines, seeking whether these mechanisms caused endogenous production of pro-inflammatory molecules or mesenchymal markers. Quantitative real-time PCR (RT-PCR) and Western blotting were used to investigate differential expression of epithelial and mesenchymal markers. Result: Incubation of iAT2 cells with LPS increased expression of IL1-ß and TNF-α in addition to mesenchymal genes, including ACTA2, FN1 and COL1A1. Alveolar epithelial cell gene expression due to LPS was significantly inhibited by GHRH-R peptide antagonist MIA-602. Incubation of iAT2 cells with cytokines like those in fibrotic lungs similarly increased expression of genes for IL1-ß, TNF-α, TGFß-1, Wnt5a, smooth muscle actin, fibronectin and collagen. Expression of mesenchymal proteins, such as N-cadherin and vimentin, were also elevated after prolonged exposure to cytokines, confirming epithelial production of pro-inflammatory molecules as an important mechanism that might lead to subsequent fibrosis. Conclusion: iAT2 cells clearly expressed the GHRH-R. Exposure to LPS or cytokines increased iAT2 cell production of pro-inflammatory factors. GHRH-R antagonist MIA-602 inhibited pro-inflammatory gene expression, implicating iAT2 cell GHRH-R signaling in lung inflammation and potentially in fibrosis.


Subject(s)
Pneumonia , Pulmonary Fibrosis , Humans , Alveolar Epithelial Cells/metabolism , Tumor Necrosis Factor-alpha , Lipopolysaccharides/pharmacology , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Inflammation , Cytokines
6.
Proc Natl Acad Sci U S A ; 116(6): 2226-2231, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30659154

ABSTRACT

Malignant pleural mesothelioma (MPM) is an aggressive malignancy associated with exposure to asbestos, with poor prognosis and no effective therapies. The strong inhibitory activities of growth hormone-releasing hormone (GHRH) antagonists have been demonstrated in different experimental human cancers, including lung cancer; however, their role in MPM remains unknown. We assessed the effects of the GHRH antagonists MIA-602 and MIA-690 in vitro in MPM cell lines and in primary MPM cells, and in vivo in MPM xenografts. GHRH, GHRH receptor, and its main splice variant SV1 were found in all the MPM cell types examined. In vitro, MIA-602 and MIA-690 reduced survival and proliferation in both MPM cell lines and primary cells and showed synergistic inhibitory activity with the chemotherapy drug pemetrexed. In MPM cells, GHRH antagonists also regulated activity and expression of apoptotic molecules, inhibited cell migration, and reduced the expression of matrix metalloproteinases. These effects were accompanied by impairment of mitochondrial activity and increased production of reactive oxygen species. In vivo, s.c. administration of MIA-602 and MIA-690 at the dose of 5 µg/d for 4 wk strongly inhibited the growth of MPM xenografts in mice, along with reduction of tumor insulin-like growth factor-I and vascular endothelial growth factor. Overall, these results suggest that treatment with GHRH antagonists, alone or in association with chemotherapy, may offer an approach for the treatment of MPM.


Subject(s)
Antineoplastic Agents/pharmacology , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mesothelioma/metabolism , Mesothelioma/pathology , Pleural Neoplasms/metabolism , Pleural Neoplasms/pathology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Gene Expression , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Humans , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Mesothelioma, Malignant , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Pleural Neoplasms/drug therapy , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Xenograft Model Antitumor Assays
7.
Molecules ; 27(9)2022 Apr 21.
Article in English | MEDLINE | ID: mdl-35566020

ABSTRACT

Antagonists of growth hormone-releasing hormone (GHRH) inhibit the growth of various tumors, including endometrial carcinomas (EC). However, tumoral receptors that mediate the antiproliferative effects of GHRH antagonists in human ECs have not been fully characterized. In this study, we investigated the expression of mRNA for GHRH and splice variants (SVs) of GHRH receptors (GHRH-R) in 39 human ECs and in 7 normal endometrial tissue samples using RT-PCR. Primers designed for the PCR amplification of mRNA for the full length GHRH-R and SVs were utilized. The PCR products were sequenced, and their specificity was confirmed. Nine ECs cancers (23%) expressed mRNA for SV1, three (7.7%) showed SV2 and eight (20.5%) revealed mRNA for SV4. The presence of SVs for GHRH-Rs could not be detected in any of the normal endometrial tissue specimens. The presence of specific, high affinity GHRH-Rs was also demonstrated in EC specimens using radioligand binding studies. Twenty-four of the investigated thirty-nine tumor samples (61.5%) and three of the seven corresponding normal endometrial tissues (42.9%) expressed mRNA for GHRH ligand. Our findings suggest the possible existence of an autocrine loop in EC based on GHRH and its tumoral SV receptors. The antiproliferative effects of GHRH antagonists on EC are likely to be exerted in part by the local SVs and GHRH system.


Subject(s)
Alternative Splicing , Endometrial Neoplasms , Growth Hormone-Releasing Hormone/genetics , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , DNA Primers , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Female , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
J Neurosci ; 40(49): 9455-9466, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33158965

ABSTRACT

Gonadal steroids modulate growth hormone (GH) secretion and the pubertal growth spurt via undefined central pathways. GH-releasing hormone (GHRH) neurons express estrogen receptor α (ERα) and androgen receptor (AR), suggesting changing levels of gonadal steroids during puberty directly modulate the somatotropic axis. We generated mice with deletion of ERα in GHRH cells (GHRHΔERα), which displayed reduced body length in both sexes. Timing of puberty onset was similar in both groups, but puberty completion was delayed in GHRHΔERα females. Lack of AR in GHRH cells (GHRHΔAR mice) induced no changes in body length, but puberty completion was also delayed in females. Using a mouse model with two reporter genes, we observed that, while GHRHtdTom neurons minimally colocalize with Kiss1hrGFP in prepubertal mice, ∼30% of GHRH neurons coexpressed both reporter genes in adult females, but not in males. Developmental analysis of Ghrh and Kiss1 expression suggested that a subpopulation of ERα neurons in the arcuate nucleus of female mice undergoes a shift in phenotype, from GHRH to Kiss1, during pubertal transition. Our findings demonstrate that direct actions of gonadal steroids in GHRH neurons modulate growth and puberty and indicate that GHRH/Kiss1 dual-phenotype neurons play a sex-specific role in the crosstalk between the somatotropic and gonadotropic axes during pubertal transition.SIGNIFICANCE STATEMENT Late maturing adolescents usually show delayed growth and bone age. At puberty, gonadal steroids have stimulatory effects on the activation of growth and reproductive axes, but the existence of gonadal steroid-sensitive neuronal crosstalk remains undefined. Moreover, the neural basis for the sex differences observed in the clinical arena is unknown. Lack of ERα in GHRH neurons disrupts growth in both sexes and causes pubertal delay in females. Deletion of androgen receptor in GHRH neurons only delayed female puberty. In adult females, not males, a subset of GHRH neurons shift phenotype to start producing Kiss1. Thus, direct estrogen action in GHRH/Kiss1 dual-phenotype neurons modulates growth and puberty and may orchestrate the sex differences in endocrine function observed during pubertal transition.


Subject(s)
Estrogen Receptor alpha/physiology , Growth Hormone-Releasing Hormone/physiology , Growth/physiology , Kisspeptins/physiology , Sexual Maturation/physiology , Signal Transduction/physiology , Animals , Estrogen Receptor alpha/genetics , Female , Gonadal Steroid Hormones/blood , Gonadal Steroid Hormones/physiology , Growth/genetics , Growth Hormone-Releasing Hormone/genetics , Hypothalamus/metabolism , Kisspeptins/genetics , Male , Mice , Mice, Knockout , Receptors, Androgen/physiology , Sex Characteristics , Sexual Maturation/genetics , Signal Transduction/genetics
9.
J Neurosci ; 40(22): 4309-4322, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32317389

ABSTRACT

Classical studies suggest that growth hormone (GH) secretion is controlled by negative-feedback loops mediated by GH-releasing hormone (GHRH)- or somatostatin-expressing neurons. Catecholamines are known to alter GH secretion and neurons expressing TH are located in several brain areas containing GH-responsive cells. However, whether TH-expressing neurons are required to regulate GH secretion via negative-feedback mechanisms is unknown. In the present study, we showed that between 50% and 90% of TH-expressing neurons in the periventricular, paraventricular, and arcuate hypothalamic nuclei and locus ceruleus of mice exhibited STAT5 phosphorylation (pSTAT5) after an acute GH injection. Ablation of GH receptor (GHR) from TH cells or in the entire brain markedly increased GH pulse secretion and body growth in both male and female mice. In contrast, GHR ablation in cells that express the dopamine transporter (DAT) or dopamine ß-hydroxylase (DBH; marker of noradrenergic/adrenergic cells) did not affect body growth. Nevertheless, less than 50% of TH-expressing neurons in the hypothalamus were found to express DAT. Ablation of GHR in TH cells increased the hypothalamic expression of Ghrh mRNA, although very few GHRH neurons were found to coexpress TH- and GH-induced pSTAT5. In summary, TH neurons that do not express DAT or DBH are required for the autoregulation of GH secretion via a negative-feedback loop. Our findings revealed a critical and previously unidentified group of catecholaminergic interneurons that are apt to sense changes in GH levels and regulate the somatotropic axis in mice.SIGNIFICANCE STATEMENT Textbooks indicate until now that the pulsatile pattern of growth hormone (GH) secretion is primarily controlled by GH-releasing hormone and somatostatin neurons. The regulation of GH secretion relies on the ability of these cells to sense changes in circulating GH levels to adjust pituitary GH secretion within a narrow physiological range. However, our study identifies a specific population of tyrosine hydroxylase-expressing neurons that is critical to autoregulate GH secretion via a negative-feedback loop. The lack of this mechanism in transgenic mice results in aberrant GH secretion and body growth. Since GH plays a key role in cell proliferation, body growth, and metabolism, our findings provide a major advance to understand how the brain regulates the somatotropic axis.


Subject(s)
Exocytosis , Feedback, Physiological , Growth Hormone/metabolism , Neurons/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine beta-Hydroxylase/genetics , Dopamine beta-Hydroxylase/metabolism , Female , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Hypothalamus/metabolism , Locus Coeruleus/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Somatotropin/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Tyrosine 3-Monooxygenase/genetics
10.
Fish Physiol Biochem ; 47(1): 69-78, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33118088

ABSTRACT

Growth hormone-releasing hormone (GHRH) is a neuropeptide that controls growth hormone (GH) synthesis and release. In this study, the full-length cDNA of Mastacembelus armatus ghrh was obtained by rapid amplification of cDNA ends method. Sequence analysis showed that the cloned sequence is 1090 bp in length, containing an open reading frame (ORF) of 429 bp that encodes a precursor protein of 142 amino acids. Sequence alignment revealed that the 27-amino acid mature peptide of Ghrh in M. armatus is conserved. Real-time PCR showed that ghrh is highly expressed in the brain, with very low or no expression in other tissues. During embryonic and larval development, ghrh expression was low in embryos but increased gradually in the stages of larval development. The biological function of Ghrh peptide was further investigated in vivo. Ghrh injection could significantly upregulate the mRNA expression of growth hormone (gh) and insulin-like growth factor-1/2 (igf-1/2) in M. armatus. Our data indicate that Ghrh is able to activate the GH-IGFs axis in M. armatus.


Subject(s)
Eels/genetics , Fish Proteins/genetics , Growth Hormone-Releasing Hormone/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , DNA, Complementary/genetics , Phylogeny
11.
Pharmacol Res ; 152: 104614, 2020 02.
Article in English | MEDLINE | ID: mdl-31874252

ABSTRACT

The somatotropic axis, in addition to its well-known metabolic and endocrine effects, plays a pivotal role in modulation of inflammation. Moreover, growth hormone (GH)-releasing hormone (GHRH) has been involved in the development of various human tumors. In this work we aimed to investigate the consequences of GHRH deficiency on the development of inflammation-associated colon carcinogenesis in a mouse model of isolated GH deficiency due to generalized ablation of the GHRH gene [GHRH knock out (GHRHKO)]. Homozygous GHRHKO (-/-) male mice and wild type (C57/BL6, +/+) male mice as control group, were used. After azoxymetane (AOM)/dextran sodium sulfate (DSS) treatment -/- mice displayed higher Disease Activity Index (DAI) score, and more marked weight loss compared to +/+ animals. Additionally, -/- mice showed a significant increase in total tumors, in particular of large size predominantly localized in distal colon. In colonic tissue of AOM/DSS-treated -/- mice we found the presence of invasive adenocarcinomas, dysplasia and colitis with mucosal ulceration. Conversely, AOM/DSS-treated +/+ mice showed only presence of adenomas, without invasion of sub-mucosa. Treatment with AOM/DSS significantly increased prostaglandin (PG)E2 and 8-iso-PGF2α levels along with cyclooxygenase-2 (COX-2), tumor necrosis factor (TNF)-α, nuclear factor kappa B (NF-kB) and inducible nitric oxide synthase (iNOS) gene expression, in colon specimens. The degree of increase of all these parameters was more markedly in -/- than +/+ mice. In conclusion, generalized GHRH ablation increases colon carcinogenesis responsiveness in male mice. Whether this results from lack of GH or GHRH remains to be established.


Subject(s)
Colitis/complications , Growth Hormone-Releasing Hormone/deficiency , Neoplasms/etiology , Adiponectin/blood , Adiponectin/genetics , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Cytokines/genetics , Dextran Sulfate , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Dinoprostone/metabolism , Growth Hormone-Releasing Hormone/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
12.
Proc Natl Acad Sci U S A ; 114(6): 1359-1364, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28123062

ABSTRACT

The etiology of benign prostatic hyperplasia (BPH) is multifactorial, and chronic inflammation plays a pivotal role in its pathogenesis. Growth hormone-releasing hormone (GHRH) is a hypothalamic neuropeptide that has been shown to act as paracrine/autocrine factor in various malignancies including prostate cancer. GHRH and its receptors are expressed in experimental models of BPH, in which antagonists of GHRH suppressed the levels of proinflammatory cytokines and altered the expression of genes related to epithelial-to-mesenchymal transition (EMT). We investigated the effects of GHRH antagonist on prostatic enlargement induced by inflammation. Autoimmune prostatitis in Balb/C mice was induced by a homogenate of reproductive tissues of male rats. During the 8-wk induction of chronic prostatitis, we detected a progressive increase in prostatic volume reaching 92% at week 8 compared with control (P < 0.001). Daily treatment for 1 mo with GHRH antagonist MIA-690 caused a 30% reduction in prostate volume (P < 0.05). Conditioned medium derived from macrophages increased the average volume of spheres by 82.7% (P < 0.001) and elevated the expression of mRNA for N-cadherin, Snail, and GHRH GHRH antagonist reduced the average volume of spheres stimulated by inflammation by 75.5% (P < 0.05), and TGF-ß2 by 91.8% (P < 0.01). The proliferation of primary epithelial cells stimulated by IL-17A or TGF-ß2 was also inhibited by 124.1% and 69.9%, respectively. GHRH stimulated the growth of BPH-1 and primary prostate spheres. This study provides evidence that GHRH plays important roles in prostatic inflammation and EMT and suggests the merit of further investigation to elucidate the effects of GHRH antagonists in prostatitis and BPH.


Subject(s)
Cell Proliferation/drug effects , Epithelial Cells/drug effects , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Cell Proliferation/genetics , Cells, Cultured , Epithelial Cells/metabolism , Gene Expression/drug effects , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Humans , Male , Mice, Inbred BALB C , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatitis/genetics , Prostatitis/metabolism , Prostatitis/pathology , Rats , Transforming Growth Factor beta2/pharmacology
13.
Cell Physiol Biochem ; 53(5): 851-864, 2019.
Article in English | MEDLINE | ID: mdl-31714043

ABSTRACT

BACKGROUND/AIMS: The growth promoting effect of lysine and betaine as well as the expression of candidate genes reflecting their efficacy, such as ghrelin, leptin, Growth Hormone Secretagogue Receptor (GHS-R), Insulin like Growth Factor (IGF- 1) and Growth Hormone Releasing Hormone (GHRH) was examined in Labeo rohita fingerlings. METHODS: One hundred eighty healthy juveniles from a homologous population were randomly distributed to 15 rectangular tanks of 150 litres capacity. The experiment was carried out for 60 days with five treatment groups consisting T1 (0.25% Betaine), T2 (0.5% Betaine), T3 (0.75% Lysine) and T4 (1.5% Lysine) and control group. The experiment was carried out for 60 days with five treatment groups consisting T1 (0.25% Betaine), T2 (0.5% Betaine), T3 (0.75% Lysine) and T4 (1.5% Lysine) and control group. At the end of trial, the growth parameters such as weight gain, SGR, PER were estimated from the weight of the triplicate groups. The digestive, metabolic and antioxidant enzymes were analysed using spectrophotometric methods. The intestine, brain and liver were sampled from the treatments and expression of different genes ghrelin, leptin, GHSR, IGF-1 and GHRH was also performed by realtime PCR. RESULTS: A significant (P<0.05) increase in weight gain, SGR, PER and lowest FCR was found in T4 group which was significantly (p < 0.05) different from other experimental groups. The highest mRNA expression levels of expression were found in T4 group which was similar to that of ghrelin gene mRNA of T2 group. The significantly (p<0.05) highest GHSR, GHRH and IGF-1 gene expression levels were found in T4 treatment group compared to other groups. CONCLUSION: The present study reveals that the lysine and betaine stimulate growth and expression of ghrelin GHRH, GHS-R and IGF-1 genes. The increase of IGF-I mRNA expression with lysine and betaine supplementation revealed that these compounds act as growth modulators. However, lysine was found to be a more potent modulator of growth compared to betaine.


Subject(s)
Betaine/pharmacology , Cyprinidae/metabolism , Gene Expression Regulation/drug effects , Lysine/pharmacology , Animal Feed , Animals , Catalase/metabolism , Cyprinidae/growth & development , Ghrelin/genetics , Ghrelin/metabolism , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Leptin/genetics , Leptin/metabolism , Liver/enzymology , Liver/metabolism , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Superoxide Dismutase/metabolism
14.
Development ; 143(20): 3763-3773, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27578785

ABSTRACT

Neurons in the hypothalamic arcuate nucleus relay and translate important cues from the periphery into the central nervous system. However, the gene regulatory program directing their development remains poorly understood. Here, we report that the LIM-homeodomain transcription factor Isl1 is expressed in several subpopulations of developing arcuate neurons and plays crucial roles in their fate specification. Mice with conditional deletion of the Isl1 gene in developing hypothalamus display severe deficits in both feeding and linear growth. Consistent with these results, their arcuate nucleus fails to express key fate markers of Isl1-expressing neurons that regulate feeding and growth. These include the orexigenic neuropeptides AgRP and NPY for specifying AgRP-neurons, the anorexigenic neuropeptide αMSH for POMC-neurons, and two growth-stimulatory peptides, growth hormone-releasing hormone (GHRH) for GHRH-neurons and somatostatin (Sst) for Sst-neurons. Finally, we show that Isl1 directly enhances the expression of AgRP by cooperating with the key orexigenic transcription factors glucocorticoid receptor and brain-specific homeobox factor. Our results identify Isl1 as a crucial transcription factor that plays essential roles in the gene regulatory program directing development of multiple arcuate neuronal subpopulations.


Subject(s)
Arcuate Nucleus of Hypothalamus/embryology , Arcuate Nucleus of Hypothalamus/metabolism , LIM-Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Female , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , LIM-Homeodomain Proteins/genetics , Mice , Pregnancy , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
15.
Prostaglandins Other Lipid Mediat ; 144: 106362, 2019 10.
Article in English | MEDLINE | ID: mdl-31301405

ABSTRACT

Growth hormone (GH) and GH-releasing hormone (GHRH), in addition to metabolic and endocrine effects, play a role in the modulation of pain and inflammation. We aimed to elucidate the consequences of GHRH deficiency on acute nociceptive stimulation and on both acute and chronic inflammatory stimuli in a mouse model of GH deficiency. Mice with generalized ablation of the GHRH gene (GHRH knock out, GHRHKO, -/-) were compared to wild type (GHRH +/+) mice. Responsiveness to acute nociceptive stimulation and to acute inflammatory stimulation was evaluated by conventional hot plate apparatus and formalin test, respectively. We also evaluated responsiveness to colonic inflammation induced both in vivo, after dextran sodium sulfate (DSS) treatment, or ex vivo, by incubating colon segments with bacterial lipopolysaccaride (LPS). Macroscopical and histological examinations were performed, prostaglandin (PG) E2 and 8-iso-PGF2α levels and cyclooxigenase (COX)-2 and tumor necrosis factor (TNF)-α gene expression were measured. Compared to controls, -/- mice showed decreased response latency during the hot plate test, and increased licking/biting time in formalin test, particularly in the second phase of inflammation. DSS treated -/- mice showed a significant increase of colonic inflammation compared to controls. Moreover DSS treatment increased PGE2 and 8-iso-PGF2α levels, along with COX-2 and TNF-α gene expression more markedly in colon specimens of -/- mice compared to controls. LPS-induced PGE2 and 8-iso-PGF2α production from colonic segments incubated ex vivo was also increased in -/- mice. Generalized GHRH gene ablation increases sensitivity to thermal pain and both acute and persistent inflammatory stimuli in male mice.


Subject(s)
Growth Hormone-Releasing Hormone/deficiency , Growth Hormone-Releasing Hormone/genetics , Pain/genetics , Animals , Cyclooxygenase 2/genetics , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Dinoprostone/metabolism , Gene Expression Regulation, Enzymologic , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Male , Mice , Mice, Knockout , Nociception , Pain/metabolism , Pain/pathology , Pain/physiopathology , Tumor Necrosis Factor-alpha/genetics
16.
Int J Cancer ; 142(11): 2394-2404, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29435973

ABSTRACT

We investigated the effects of novel antagonists of growth hormone releasing hormone (GHRH)-MIA602 and MIA690-on three human small cell lung cancer (SCLC) lines (H446, DMS53 and H69) and two non-SCLC (NSCLC) lines (HCC827 and H460). In vitro exposure of cancer cells to these GHRH antagonists significantly inhibited cell viability, increased cell apoptosis, decrease cellular levels of cAMP and reduced cell migration. In vivo, the antagonists strongly inhibited tumor growth in xenografted nude mice models. Subcutaneous administration of MIA602 at the dose of 5 µg/day for 4-8 weeks reduced the growth of HCC827, H460 and H446 tumors by 69.9%, 68.3% and 53.4%, respectively, while MIA690 caused a reduction of 76.8%, 58.3% and 54.9%, respectively. Western blot and qRT-PCR analyses demonstrated a downregulation of expression of the pituitary-type GHRH-R and its splice-variant, cyclinD1/2, cyclin-dependent kinase4/6, p21-activated kinase-1, phosphorylation of activator of transcription 3 and cAMP response element binding protein; and an upregulation of expression of E-cadherin, ß-catenin and P27kip1 in cancer cells and in xenografted tumor tissues. The study demonstrates the involvement of GHRH antagonists in multiple signaling pathways in lung cancers. Our findings suggest the merit of further investigation with these GHRH antagonists on the management of both SCLC and NSCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Sermorelin/analogs & derivatives , Small Cell Lung Carcinoma/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Female , Gene Expression , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Humans , Mice , Mice, Nude , STAT3 Transcription Factor/metabolism , Sermorelin/pharmacology , Signal Transduction/drug effects , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Xenograft Model Antitumor Assays
17.
Cell Tissue Res ; 374(2): 349-365, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29934855

ABSTRACT

Ghrelin, a gut-brain peptide hormone, is implicated in a multiplicity of biological functions, including energy homeostasis and reproduction. Neuronal systems that are involved in energy homeostasis as well as reproduction traverse the hypothalamus; however, the mechanism by which they control energy homeostasis is not fully understood. The present study analyzes the anatomical relationship of neurons expressing gonadotropin-releasing hormone (GnRH), neuropeptide Y (NPY) and growth hormone-releasing hormone (GHRH) in a cichlid, tilapia (Oreochromis niloticus). Additionally, we examine in vivo effects of ghrelin on these hypothalamic neurons and plasma growth hormone (GH) and insulin-like growth factor-1 (IGF-1) levels. Double-immunofluorescence showed neuronal fiber associations between GnRH, NPY and GHRH in the brain and pituitary. Intracerebroventricular injection of ghrelin had no effect on numbers, soma size, or optical density of GnRH and NPY neurons, whereas the number of GHRH neurons was significantly decreased in the animals injected with ghrelin when compared to controls, which may indicate administered ghrelin promoted GHRH release. Plasma GH and pituitary GH mRNA levels were significantly increased in the animals injected with ghrelin. These results suggest that central administration of ghrelin primarily act on hypothalamic GHRH neurons to stimulate GH release from the pituitary in the tilapia.


Subject(s)
Cichlids/metabolism , Ghrelin/pharmacology , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Pituitary Gland/metabolism , Animals , Female , Ghrelin/administration & dosage , Gonadotropin-Releasing Hormone/metabolism , Growth Hormone/blood , Growth Hormone/genetics , Growth Hormone-Releasing Hormone/genetics , Humans , Hypothalamus/drug effects , Insulin-Like Growth Factor I/metabolism , Neurons/drug effects , Neuropeptide Y/metabolism , Pituitary Gland/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
18.
Gen Comp Endocrinol ; 255: 90-101, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28974369

ABSTRACT

The somatotropic axis (SA) regulates numerous aspects of vertebrate physiology such as development, growth, and metabolism and has influence on several tissues including neural, immune, reproductive and gastric tract. Growth hormone (GH) is a key component of SA, it is synthesized and released mainly by pituitary somatotrophs, although now it is known that virtually all tissues can express GH, which, in addition to its well-described endocrine roles, also has autocrine/paracrine/intracrine actions. In the pituitary, GH expression is regulated by several hypothalamic neuropeptides including GHRH, PACAP, TRH and SST. GH, in turn, regulates IGF1 synthesis in several target tissues, adding complexity to the system since GH effects can be exerted either directly or mediated by IGF1. In reptiles, little is known about the SA components and their functional interactions. The aim of this work was to characterize the mRNAs of the principal SA components in the green iguana and to develop the tools that allow the study of the structural and functional evolution of this system in reptiles. By employing RT-PCR and RACE, the cDNAs encoding for GHRH, PACAP, TRH, SST and IGF1 were amplified and sequenced. Results showed that these cDNAs coded for the corresponding protein precursors of 154, 170, 243, 113, and 131 amino acids, respectively. Of these, GHRH, PACAP, SST and IGF1 precursors exhibited a high structural conservation with respect to its counterparts in other vertebrates. On the other hand, iguana's TRH precursor showed 7 functional copies of mature TRH (pyr-QHP-NH2), as compared to 4 and 6 copies of TRH in avian and mammalian proTRH sequences, respectively. It was found that in addition to its primary production site (brain for GHRH, PACAP, TRH and SST, and liver for IGF1), they were also expressed in other peripheral tissues, i.e. testes and ovaries expressed all the studied mRNAs, whereas TRH and IGF1 mRNAs were observed ubiquitously in all tissues considered. These results show that the main SA components in reptiles of the Squamata Order maintain a good structural conservation among vertebrate phylogeny, and suggest important physiological interactions (endocrine, autocrine and/or paracrine) between them due to their wide peripheral tissue expression.


Subject(s)
Growth Hormone-Releasing Hormone/genetics , Iguanas/genetics , Insulin-Like Growth Factor I/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Somatostatin/genetics , Thyrotropin-Releasing Hormone/genetics , Amino Acid Sequence , Animals , Base Sequence , Growth Hormone-Releasing Hormone/chemistry , Growth Hormone-Releasing Hormone/metabolism , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor I/metabolism , Phylogeny , Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Somatostatin/chemistry , Somatostatin/metabolism , Thyrotropin-Releasing Hormone/chemistry , Thyrotropin-Releasing Hormone/metabolism
19.
J Physiol ; 594(24): 7309-7326, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27558671

ABSTRACT

KEY POINTS: Loss of function of the melanocortin 4 receptor (MC4R) results in hyperphagia, obesity and increased growth. Despite knowing that MC4Rs control food intake, we are yet to understand why defects in the function of the MC4R receptor contribute to rapid linear growth. We show that hyperphagia following germline loss of MC4R in male mice promotes growth while suppressing the growth hormone-insulin-like growth factor-1 (GH-IGF-1) axis. We propose that hyperinsulinaemia promotes growth while suppressing the GH-IGF-1 axis. It is argued that physiological responses essential to maintain energy flux override conventional mechanisms of pubertal growth to promote the storage of excess energy while ensuring growth. ABSTRACT: Defects in melanocortin-4-receptor (MC4R) signalling result in hyperphagia, obesity and increased growth. Clinical observations suggest that loss of MC4R function may enhance growth hormone (GH)-mediated growth, although this remains untested. Using male mice with germline loss of the MC4R, we assessed pulsatile GH release and insulin-like growth factor-1 (IGF-1) production and/or release relative to pubertal growth. We demonstrate early-onset suppression of GH release in rapidly growing MC4R deficient (MC4RKO) mice, confirming that increased linear growth in MC4RKO mice does not occur in response to enhanced activation of the GH-IGF-1 axis. The progressive suppression of GH release in MC4RKO mice occurred alongside increased adiposity and the progressive worsening of hyperphagia-associated hyperinsulinaemia. We next prevented hyperphagia in MC4RKO mice through restricting calorie intake in these mice to match that of wild-type (WT) littermates. Pair feeding of MC4RKO mice did not prevent increased adiposity, but attenuated hyperinsulinaemia, recovered GH release, and normalized linear growth rate to that seen in pair-fed WT littermate controls. We conclude that the suppression of GH release in MC4RKO mice occurs independently of increased adipose mass, and is a consequence of hyperphagia-associated hyperinsulinaemia. It is proposed that physiological responses essential to maintain energy flux (hyperinsulinaemia and the suppression of GH release) override conventional mechanisms of pubertal growth to promote the storage of excess energy while ensuring growth. Implications of these findings are likely to extend beyond individuals with defects in MC4R signalling, encompassing physiological changes central to mechanisms of growth and energy homeostasis universal to hyperphagia-associated childhood-onset obesity.


Subject(s)
Growth Hormone/metabolism , Hyperphagia/metabolism , Insulin-Like Growth Factor I/metabolism , Obesity/metabolism , Receptor, Melanocortin, Type 4/metabolism , Animals , Brain/metabolism , Green Fluorescent Proteins/genetics , Growth Hormone/genetics , Growth Hormone-Releasing Hormone/genetics , Insulin/blood , Leptin/blood , Male , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/metabolism , Neurons/metabolism , Receptor, Melanocortin, Type 4/deficiency , Receptor, Melanocortin, Type 4/genetics
20.
Mol Biol Rep ; 43(6): 517-26, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27075656

ABSTRACT

Cathepsins, growth hormone-releasing hormone (GHRH) and leptin receptor (LEPR) genes have been receiving increasing attention as potential markers for meat quality and pig performance traits. This study investigated the allele variants in four cathepsin genes (CTSB, CTSK, CTSL, CTSS), GHRH and LEPR in pure-bred Ukrainian Large White pigs and evaluated effects of the allele variants on meat quality characteristics. The study was conducted on 72 pigs. Genotyping was performed using PCR-RFLP technique. Meat quality characteristics analysed were intramuscular fat content, tenderness, total water content, ultimate pH, crude protein and ashes. A medium level of heterozygosity values was established for GHRH and LEPR genes which corresponded to very high levels of informativeness indexes. Cathepsins CTSL, CTSB and CTSK had a low level of heterozygosity, and CTSS did not segregate in this breed. Association studies established that intramuscular fat content and tenderness were affected by the allele variance in GHRH and LEPR but not by CTSB and CTSL genes. The GHRH results could be particularly relevant for the production of lean prime cuts as the A allele is associated with both, a lower meat fat content and better tenderness values, which are two attributes highly regarded by consumers. Results of this study suggest that selective breeding towards GHRH/AA genotype would be particularly useful for improving meat quality characteristics in the production systems involving lean Large White lines, which typically have less than 2 % intramuscular fat content.


Subject(s)
Cathepsins/genetics , Growth Hormone-Releasing Hormone/genetics , Meat/standards , Receptors, Leptin/genetics , Adiposity , Animals , Female , Food Quality , Genetic Association Studies , Male , Muscle, Skeletal/anatomy & histology , Polymorphism, Single Nucleotide , Sus scrofa/genetics
SELECTION OF CITATIONS
SEARCH DETAIL