1
|
Zeng M, Zhan C, Li Y, Liao H, Liu W, Chen G, Wang J. Melatonin prevents the transgenerational toxicity of nanoplastics in zebrafish (Danio rerio). THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 953:176043. [PMID: 39241878 DOI: 10.1016/j.scitotenv.2024.176043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 08/25/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
As a novel pollutant, microplastic pollution has become a global environmental concern. Melatonin (MT) has a protective effect on the damage caused by pollutants. However, there is still a lack of research on the transgenerational toxicity of microplastics and the alleviation of microplastics toxicity by MT. In this study, the adult zebrafish was exposed to (0, 0.1 and 1 mg/L) polystyrene nanoplastics (PSNP) with or without (1 μM) MT for 14 days, and embryos (F1) were used for experiments. Our study found that long-term exposure of parents to 1 mg/L PSNP reduced fertilization rate and survival rate of offspring, increased the deformity rate and induced embryos to hatch in advance. The growth inhibition of offspring was related to the gene transcription of the growth hormone/insulin-like growth factor axis. Moreover, PSNP caused oxidative stress in offspring, damaged immune system, reduced antioxidant capacity and induced apoptosis. MT supplementation could effectively alleviate the developmental toxicity and oxidative damage of offspring, but the negative effects brought by PSNP could not be completely eliminated. Our research provided a new reference for the protective effect of MT on transgenerational toxicity induced by PSNP.
Collapse
Affiliation(s)
- Min Zeng
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Chunhua Zhan
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang, Hunan 421001, China
| | - Ye Li
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Hongping Liao
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Wanjing Liu
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, University of South China, Hengyang, Hunan 421001, China
| | - Guanglong Chen
- Institute of Eco-Environmental Research, Guangxi Academy of Sciences, Nanning 530007, China.
| | - Jun Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou 511464, China; Institute of Eco-Environmental Research, Guangxi Academy of Sciences, Nanning 530007, China.
| |
Collapse
|
2
|
Juhász É, Szabó Z, Schally AV, Király J, Fodor P, Kónya G, Dezső B, Szabó E, Halmos G, Kiss C. Expression of Growth Hormone-Releasing Hormone and Its Receptor Splice Variants in a Cohort of Hungarian Pediatric Patients with Hematological and Oncological Disorders: A Pilot Study. Int J Mol Sci 2024; 25:8831. [PMID: 39201517 PMCID: PMC11354965 DOI: 10.3390/ijms25168831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Hematological and oncological diseases are still among the leading causes of childhood mortality. Expression of growth hormone-releasing hormone (GHRH) and its receptors (GHRH-R) has been previously demonstrated in various human tumors, but very limited findings are available about the presence and potential function of GHRH-Rs in oncological and hematological disorders of children. In this study, we aimed to investigate the expression of mRNA for GHRH and splice variant 1 (SV) of GHRH-R in 15 pediatric hematological/oncological specimens by RT-PCR. The presence and binding characteristics of GHRH-R protein were also studied by Western blot and ligand competition assays. Of the fifteen specimens studied, eleven pediatric samples (73%) showed the expression of mRNA for GHRH. These eleven samples also expressed mRNA for GHRH receptor SV1. GHRH-R protein was found to be expressed in two benign tumor samples and five malignant tumors examined by Western blot. The presence of specific, high affinity binding sites on GHRH-R was demonstrated in all of the seven human pediatric solid tumor samples investigated. Our results show that the expression of GHRH and SV1 of GHRH-R in hemato-oncological diseases in children can pave the way for further investigation of GHRH-Rs as potential molecular targets for diagnosis and therapy.
Collapse
Affiliation(s)
- Éva Juhász
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Zsuzsanna Szabó
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (J.K.); (P.F.); (G.K.)
| | - Andrew V. Schally
- Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL 33125, USA;
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
- Department of Medicine, Divisions of Hematology-Oncology and Endocrinology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - József Király
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (J.K.); (P.F.); (G.K.)
| | - Petra Fodor
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (J.K.); (P.F.); (G.K.)
| | - Gábor Kónya
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (J.K.); (P.F.); (G.K.)
| | - Balázs Dezső
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Erzsébet Szabó
- Department of Pharmacology, Faculty of Pharmacy, HUN-REN-DE Pharmamodul Research Group, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary;
| | - Gábor Halmos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (J.K.); (P.F.); (G.K.)
- Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL 33125, USA;
| | - Csongor Kiss
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| |
Collapse
|
3
|
Saleh AA, Hassan TGM, El-Hedainy DKA, El-Barbary ASA, Sharaby MA, Hafez EE, Rashad AMA. IGF-I and GH Genes polymorphism and their association with milk yields, composition and reproductive performance in Holstein-Friesian dairy cattle. BMC Vet Res 2024; 20:341. [PMID: 39095829 PMCID: PMC11295711 DOI: 10.1186/s12917-024-04188-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 07/11/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The insulin-like growth factor (IGF-I) and growth hormone (GH) genes have been identified as major regulators of milk yield and composition, and reproductive performance in cattle. Genetic variations/polymorphism in these genes have been found to influence milk production, yield and quality. This investigation aimed to explore the association between IGF-I and GH polymorphisms and milk yield and composition, and reproductive performance in a herd consisting of 1000 Holstein-Friesian (HF) dairy cattle from El-Alamia farm. The experimental animals were 76 ± 7.25 months in age, with an average live weight of 750 ± 50.49 kg, and raised under the same conditions of feeding and weather. The studied animals were divided into three categories; high producers (n = 280), medium producers (n = 318) and low producers (n = 402). RESULTS The digestion of 249 bp for IGF-I-SnaBI using the Restriction-fragment-length-polymorphism (RFLP) technique yielded two alleles; T (0.59) and C (0.41) and three genotypes; TT (0.52), TC (0.39) and CC (0.09) and this agrees with the results of DNA/gene sequencing technique. The sequencing analysis of the IGF-I gene revealed polymorphism in position 472 (C > T). Nucleotide sequencing of the amplified fragment of the IGF-I gene of different genotypes was done and submitted to the NCBI GenBank with Accession no. MH156812.1 and MH156811.1. While the digestion of 432 bp for GH-AluI using the RFLP technique yielded two alleles; A (0.81) and G (0.19) and two genotypes; AA (0.77) and AG (0.23) and this agrees with the results of DNA/gene sequencing technique. The sequencing analysis of the GH gene revealed polymorphism in the position 1758 C > G and in turn led to changes in amino acid sequence as Alanine for (A) compared to Glycine for (G). Nucleotide sequencing of the amplified fragment of the GH gene was done and submitted to the NCBI GenBank with Accession no. MH156810.1. The results of this study demonstrate the effects of variants of the GH-IGF-I somatotrophic axis on milk production and composition traits in commercial HF cattle. The greatest values of milk yield and reproductive performance were observed on IGF-I-SnaBI-TC and GH-AluI-AG genotypes. While the greatest % fat and % protein values were observed on IGF-I-SnaBI-CC and GH-AluI-AA genotyped individuals. CONCLUSION The genetic variation of the studied genes can be utilized in selecting animals with superior milk yield, composition and reproductive performance in Holstein-Friesian Dairy Cattle under subtropical conditions.
Collapse
Affiliation(s)
- Ahmed A Saleh
- Animal and Fish Production Department, Faculty of Agriculture (Al-Shatby), Alexandria University, Aflaton St, Alexandria City, 11865, Egypt.
| | - Tarek G M Hassan
- Animal and Fish Production Department, Faculty of Agriculture (Al-Shatby), Alexandria University, Aflaton St, Alexandria City, 11865, Egypt
| | - Dalia K A El-Hedainy
- Animal and Fish Production Department, Faculty of Agriculture (Al-Shatby), Alexandria University, Aflaton St, Alexandria City, 11865, Egypt
| | - Adel S A El-Barbary
- Animal and Fish Production Department, Faculty of Agriculture (Al-Shatby), Alexandria University, Aflaton St, Alexandria City, 11865, Egypt
| | - Mahmoud A Sharaby
- Animal and Fish Production Department, Faculty of Agriculture (Al-Shatby), Alexandria University, Aflaton St, Alexandria City, 11865, Egypt
| | - Elsayed E Hafez
- Arid Lands Cultivation Research Institute, City of Scientific Research and Technological Applications, Alexandria, New Borg El Arab, 21934, Egypt
| | - Amr M A Rashad
- Animal and Fish Production Department, Faculty of Agriculture (Al-Shatby), Alexandria University, Aflaton St, Alexandria City, 11865, Egypt
| |
Collapse
|
4
|
Wei S, Zhang M, Li Y, Yang W, Zhang C, Liu F, Chen S, Ban B, He D. Identification and functional analysis of first heterozygous frameshift mutation in the GHRH gene in a Chinese boy with isolated growth hormone deficiency. Gene 2024; 907:148283. [PMID: 38354915 DOI: 10.1016/j.gene.2024.148283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/26/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Isolated growth hormone deficiency (IGHD) is a rare genetically heterogeneous disorder caused primarily by mutations in GH1 and GH releasing hormone receptor (GHRHR). The aim of this study was to identify the molecular etiology of a Chinese boy with IGHD. METHODS Whole-exome sequencing, sanger sequencing and bioinformatic analysis were performed to screen for candidate mutations. The impacts of candidate mutation on gene expression, intracellular localization and protein function were further evaluated by in vitro assays. RESULTS A novel heterozygous frameshift mutation in the GHRH gene (c.91dupC, p.R31Pfs*98) was identified in a Chinese boy clinically diagnosed as having IGHD. The mutation was absent in multiple public databases, and considered as deleterious using in silico prediction, conservative analysis and three-dimensional homology modeling. Furthermore, mRNA and protein expression levels of mutant GHRH were significantly increased than wild-type GHRH (p < 0.05). Moreover, mutant GHRH showed an aberrant accumulation within the cytoplasm, and obviously reduced ability to stimulate GH secretion and cAMP accumulation in human GHRHR-expressing pituitary GH3 cells compared to wild-type GHRH (p < 0.05). CONCLUSION Our study discovered the first loss-of function mutation of GHRH in a Chinese boy with IGHD and provided new insights on IGHD pathogenesis caused by GHRH haploinsufficiency.
Collapse
Affiliation(s)
- Shuoshuo Wei
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, PR China; Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, PR China
| | - Mei Zhang
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, PR China; Chinese Research Center for Behavior Medicine in Growth and Development, Jining, PR China
| | - Yanying Li
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, PR China; Chinese Research Center for Behavior Medicine in Growth and Development, Jining, PR China
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, PR China
| | - Chuanpeng Zhang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, PR China
| | - Fupeng Liu
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, PR China; Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, PR China
| | - Shuxiong Chen
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, PR China; Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, PR China
| | - Bo Ban
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, PR China; Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, PR China; Chinese Research Center for Behavior Medicine in Growth and Development, Jining, PR China.
| | - Dongye He
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, PR China; Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, PR China.
| |
Collapse
|
5
|
Halmos G, Szabo Z, Juhasz E, Schally AV. Signaling mechanism of growth hormone-releasing hormone receptor. VITAMINS AND HORMONES 2023; 123:1-26. [PMID: 37717982 DOI: 10.1016/bs.vh.2023.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
The hypothalamic peptide growth hormone-releasing hormone (GHRH) stimulates the secretion of growth hormone (GH) from the pituitary through binding and activation of the pituitary type of GHRH receptor (GHRH-R), which belongs to the family of G protein-coupled receptors with seven potential membrane-spanning domains. Splice variants of GHRH-Rs (SV) in human tumors and other extra pituitary tissues were identified and their cDNA was sequenced. Among the SVs, splice variant 1 (SV1) possesses the greatest similarity to the full-length GHRH-R and remains functional by eliciting cAMP signaling and mitogenic activity upon GHRH stimulation. A large body of work have evaluated potential clinical applications of agonists and antagonists of GHRH in diverse fields, including endocrinology, oncology, cardiology, diabetes, obesity, metabolic dysfunctions, Alzheimer's disease, ophthalmology, wound healing and other applications. In this chapter, we briefly review the expression and potential function of GHRH-Rs and their SVs in various tissues and also elucidate and summarize the activation, molecular mechanism and signalization pathways of these receptors. Therapeutic applications of GHRH analogs are also discussed.
Collapse
Affiliation(s)
- Gabor Halmos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL, United States.
| | - Zsuzsanna Szabo
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Eva Juhasz
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andrew V Schally
- Veterans Affairs Medical Center, Endocrine, Polypeptide and Cancer Institute, Miami, FL, United States; Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL, United States; Department of Medicine, Divisions of Hematology-Oncology and Endocrinology, Miller School of Medicine, University of Miami, Miami, FL, United States; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States
| |
Collapse
|
6
|
Polymorphisms of the Growth Hormone Releasing Hormone Receptor Gene Affect Body Conformation Traits in Chinese Dabieshan Cattle. Animals (Basel) 2022; 12:ani12131601. [PMID: 35804502 PMCID: PMC9265053 DOI: 10.3390/ani12131601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022] Open
Abstract
This study was performed to expose the polymorphisms of the growth hormone-releasing hormone receptor gene in Chinese Dabieshan cattle, evaluate its effect on body conformation traits, and find potential molecular markers in Chinese cattle. The GHRHR structure and the phylogenetic tree were analyzed using bioinformatics software. The polymorphism of the GHRHR gene in 486 female cattle was genotyped by PCR-RFLP and DNA sequencing, and the association between SNPs and body conformation traits of Chinese Dabieshan cattle was analyzed by one-way ANOVA in SPSS software. GHRHR was often conserved in nine species, and its sequence of cattle was closest to sheep and goats. Six polymorphic SNPs were identified, g.10667A > C and g.10670A > C were missense mutation. The association analysis indicated that the six SNPs significantly influenced the body conformation traits of Chinese Dabieshan cattle (p < 0.05). Six haplotypes were identified and Hap1 (-CAACGA-) had the highest frequency (36.10%). The Hap3/5 (-GCCCCCGGAAGG-) exhibited a significantly greater wither height (WH), hip height (HH), heart girth (HG), and hip width (HW) (p < 0.05). Overall, the polymorphisms of GHRHR affected the body conformation traits of Chinese Dabieshan cattle, and the GHRHR gene could be used as a molecular marker in Dabieshan cattle breeding programs.
Collapse
|
7
|
Teng Z, Hao L, Yang R, Song J, Wang Z, Jiao Y, Fang J, Zheng S, Ma Z, Chen X, Liu S, Cheng Y. Key pituitary miRNAs mediate the expression of pig GHRHR splice variants by regulating splice factors. Int J Biol Macromol 2022; 208:208-218. [PMID: 35306020 DOI: 10.1016/j.ijbiomac.2022.03.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 11/28/2022]
Abstract
The growth hormone releasing hormone receptor (GHRHR) is well documented in organism growth and its alternative splicing may generate multiple functional GHRHR splice variants (SVs). Our previous study has demonstrated the key pituitary miRNAs (let-7e and miR-328-5p) in pig regulated the expression of GHRHR SVs by directly targeting to them. And according to recent reports, the interplay between miRNA-based silencing of mRNAs and alternative splicing of pre-mRNAs is a crucial post-transcriptional mechanism. In this study, SF3B3 and CPSF4 were firstly excavated as the splice factors that involved in the formation of GHRHR SVs mediated by let-7e and miR-328-5p through the comparation of the expression relations of GHRHR SVs, let-7e/miR-328-5p and SF3B3/CPSF4 in pituitary tissues between Landrace pigs and BaMa pigs, as well as the prediction of the target relations of let-7e/miR-328-5p with SF3B3 and/or CPSF4. SF3B3 and CPSF4 targeted by let-7e and miR-328-5p were further verified by performing dual-luciferase reporter assays and detecting the expression of target transcripts. Then the RT-PCR, RT-qPCR and Western blot assays were used to confirm SF3B3 and CPSF4 were involved in the formation of the GHRHR SVs, and in this process, let-7e and miR-328-5p mediated GHRHR SVs by regulating SF3B3 and CPSF4. Finally, the target site of SF3B3 on pre-GHRHR was on the Exon 12 to Exon14, while CPSF4 acted on the other fragments of the pre-GHRHR, which were explored by dual-luciferase reporter system preliminarily. To the best of our knowledge, this paper is the first to report the miRNAs regulate GHRHR SVs indirectly by splice factors.
Collapse
Affiliation(s)
- Zhaohui Teng
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Rui Yang
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Jie Song
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Zhaoguo Wang
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Yingying Jiao
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Jiayuan Fang
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Shuo Zheng
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Ze Ma
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Xi Chen
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Songcai Liu
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Yunyun Cheng
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
8
|
Sato T, Ida T, Shiimura Y, Matsui K, Oishi K, Kojima M. Insights Into the Regulation of Offspring Growth by Maternally Derived Ghrelin. Front Endocrinol (Lausanne) 2022; 13:852636. [PMID: 35250893 PMCID: PMC8894672 DOI: 10.3389/fendo.2022.852636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
The regulation of fetal development by bioactive substances such as hormones and neuropeptides derived from the gestational mother is considered to be essential for the development of the fetus. On the other hand, it has been suggested that changes in the physiological state of the pregnant mother due to various factors may alter the secretion of these bioactive substances and induce metabolic changes in the offspring, such as obesity, overeating, and inflammation, thereby affecting postnatal growth and health. However, our knowledge of how gestational maternal bioactive substances modulate offspring physiology remains fragmented and lacks a systematic understanding. In this mini-review, we focus on ghrelin, which regulates growth and energy metabolism, to advance our understanding of the mechanisms by which maternally derived ghrelin regulates the growth and health of the offspring. Understanding the regulation of offspring growth by maternally-derived ghrelin is expected to clarify the fetal onset of metabolic abnormalities and lead to a better understanding of lifelong health in the next generation of offspring.
Collapse
Affiliation(s)
- Takahiro Sato
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Japan
- *Correspondence: Takahiro Sato, ; Masayasu Kojima,
| | - Takanori Ida
- Division for Identification and Analysis of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Yuki Shiimura
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Japan
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Kazuma Matsui
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Japan
| | - Kanae Oishi
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Japan
| | - Masayasu Kojima
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Japan
- *Correspondence: Takahiro Sato, ; Masayasu Kojima,
| |
Collapse
|
9
|
Scalco RC, Correa FA, Dantas NCB, Vasques GA, Jorge AAL. Hormone resistance and short stature: A journey through the pathways of hormone signaling. Mol Cell Endocrinol 2021; 536:111416. [PMID: 34333056 DOI: 10.1016/j.mce.2021.111416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 11/20/2022]
Abstract
Hormone resistances have been described in association with growth disorders, the majority involving the growth hormone (GH)/insulin-like growth factor 1(IGF-1) axis or hormones with specific paracrine-autocrine actions in the growth plate. Defects in hormone receptors or in proteins involved in intracellular signal transduction (post-receptor defects) are the main mechanisms of hormone resistance leading to short stature. The characteristic phenotypes of each of these hormonal resistances are very distinct and bring with them important insights into the role of each hormone and its signaling pathway. In this review, we discuss the molecular and clinical aspects of the main hormone resistances associated with short stature in humans.
Collapse
Affiliation(s)
- Renata C Scalco
- Disciplina de Endocrinologia, Faculdade de Ciencias Medicas da Santa Casa de Sao Paulo, Brazil
| | - Fernanda A Correa
- Unidade de Endocrinologia do Desenvolvimento, Laboratorio de Hormonios e Genetica Molecular (LIM/42) do Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo (HC-FMUSP), Brazil; Instituto do Cancer do Estado de Sao Paulo (ICESP) da Faculdade de Medicina da Universidade de São Paulo (FMUSP), Brazil
| | - Naiara C B Dantas
- Unidade de Endocrinologia do Desenvolvimento, Laboratorio de Hormonios e Genetica Molecular (LIM/42) do Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo (HC-FMUSP), Brazil; Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular (LIM/25) do Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo (HC-FMUSP), Brazil
| | - Gabriela A Vasques
- Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular (LIM/25) do Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo (HC-FMUSP), Brazil
| | - Alexander A L Jorge
- Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular (LIM/25) do Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo (HC-FMUSP), Brazil.
| |
Collapse
|
10
|
Cen LP, Ng TK, Liang JJ, Xu C, Zhuang X, Liu YF, Chen SL, Xu Y, Yang Q, Yuan XL, Qin YJ, Chan SO, Chen H, Zhang M, Schally AV, Pang CP. Agonist of growth hormone-releasing hormone enhances retinal ganglion cell protection induced by macrophages after optic nerve injury. Proc Natl Acad Sci U S A 2021; 118:e1920834118. [PMID: 34244423 PMCID: PMC8285901 DOI: 10.1073/pnas.1920834118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Optic neuropathies are leading causes of irreversible visual impairment and blindness, currently affecting more than 100 million people worldwide. Glaucoma is a group of optic neuropathies attributed to progressive degeneration of retinal ganglion cells (RGCs). We have previously demonstrated an increase in survival of RGCs by the activation of macrophages, whereas the inhibition of macrophages was involved in the alleviation on endotoxin-induced inflammation by antagonist of growth hormone-releasing hormone (GHRH). Herein, we hypothesized that GHRH receptor (GHRH-R) signaling could be involved in the survival of RGCs mediated by inflammation. We found the expression of GHRH-R in RGCs of adult rat retina. After optic nerve crush, subcutaneous application of GHRH agonist MR-409 or antagonist MIA-602 promoted the survival of RGCs. Both the GHRH agonist and antagonist increased the phosphorylation of Akt in the retina, but only agonist MR-409 promoted microglia activation in the retina. The antagonist MIA-602 reduced significantly the expression of inflammation-related genes Il1b, Il6, and Tnf Moreover, agonist MR-409 further enhanced the promotion of RGC survival by lens injury or zymosan-induced macrophage activation, whereas antagonist MIA-602 attenuated the enhancement in RGC survival. Our findings reveal the protective effect of agonistic analogs of GHRH on RGCs in rats after optic nerve injury and its additive effect to macrophage activation, indicating a therapeutic potential of GHRH agonists for the protection of RGCs against optic neuropathies especially in glaucoma.
Collapse
Affiliation(s)
- Ling-Ping Cen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
- Shantou University Medical College, 515041 Shantou, China
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Jia-Jian Liang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
| | - Ciyan Xu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
| | - Xi Zhuang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
| | - Yu-Fen Liu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
- Shantou University Medical College, 515041 Shantou, China
| | - Shao-Lang Chen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
| | - Yanxuan Xu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
| | - Qichen Yang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Xiang-Ling Yuan
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
- Shantou University Medical College, 515041 Shantou, China
| | - Yong Jie Qin
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong General Hospital and Guangdong Academy of Medical Sciences, 510080 Guangzhou, China
| | - Sun On Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Haoyu Chen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
| | - Mingzhi Zhang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China
| | - Andrew V Schally
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136;
- Division of Medical Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136
- Division of Endocrinology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136
- Cancer Institute, Veterans Affairs Medical Center, Miami, FL 33125
| | - Chi Pui Pang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, 515041 Shantou, China;
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
11
|
Zhang Y, Yi X, Huang K, Sun Q, Kong R, Chen S, Liang C, Li M, Letcher RJ, Liu C. Tris(1,3-dichloro-2-propyl)phosphate Reduces Growth Hormone Expression via Binding to Growth Hormone Releasing Hormone Receptors and Inhibits the Growth of Crucian Carp. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:8108-8118. [PMID: 34062063 DOI: 10.1021/acs.est.0c07708] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Tris(1,3-dichloro-2-propyl)phosphate (TDCIPP) has commonly been used as an additive flame retardant and frequently detected in the aquatic environment and in biological samples worldwide. Recently, it was found that exposure to TDCIPP inhibited the growth of zebrafish, but the relevant molecular mechanisms remained unclear. In this study, 5 day-old crucian carp (Carassius auratus) larvae were treated with 0.5, 5, or 50 μg/L TDCIPP for 90 days; the effect on growth was evaluated; and related molecular mechanisms were explored. Results demonstrated that 5 or 50 μg/L TDCIPP treatment significantly inhibited the growth of crucian carp and downregulated the expression of growth hormones (ghs), growth hormone receptor (ghr), and insulin-like growth factor 1 (igf1). Molecular docking, dual-luciferase reporter gene assay, and in vitro experiments demonstrated that TDCIPP could bind to the growth hormone releasing hormone receptor protein of crucian carp and disturb the stimulation of growth hormone releasing hormone to the expression of ghs, resulting in the decrease of the mRNA level of gh1 and gh2 in pituitary cells. Our findings provide new perceptions into the molecular mechanisms of developmental toxicity of TDCIPP in fish.
Collapse
Affiliation(s)
- Yongkang Zhang
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xun'e Yi
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Kai Huang
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qian Sun
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Ren Kong
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Sheng Chen
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Chengqian Liang
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Meng Li
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Robert J Letcher
- Departments of Chemistry and Biology, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Chunsheng Liu
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
12
|
Omouessi ST, Leipprandt JR, Akoume MY, Charbeneau R, Wade S, Neubig RR. Mice with an RGS-insensitive Gα i2 protein show growth hormone axis dysfunction. Mol Cell Endocrinol 2021; 521:111098. [PMID: 33278490 DOI: 10.1016/j.mce.2020.111098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/22/2020] [Accepted: 11/26/2020] [Indexed: 01/03/2023]
Abstract
Mice carrying an RGS-insensitive Gαi2 mutation display growth retardation early after birth. Although the growth hormone (GH)-axis is a key endocrine modulator of postnatal growth, its functional state in these mice has not been characterized. The present study was undertaken to address this issue. Results revealed that pituitary mRNA levels for GH, prolactin (PRL), somatostatin (SST), GH-releasing-hormone receptor (GHRH-R) and GH secretagogue receptor (GHS-R) were decreased in mutants compared to controls. These changes were reflected by a significant decrease in plasma levels of GH, IGF-1 and IGF-binding protein-3 (IGFBP-3). Mutants were also less responsive to GHRH and ghrelin (GhL) on GH stimulation of release from pituitary primary cell cultures. In contrast, they were more sensitive to the inhibitory effect of SST. These data provide the first evidence for an alteration of the functional state of the GH-axis in Gαi2G184S mice that likely contributes to their growth retardation.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Female
- GTP-Binding Protein alpha Subunit, Gi2/genetics
- GTP-Binding Protein alpha Subunit, Gi2/metabolism
- Ghrelin/pharmacology
- Growth Disorders/genetics
- Growth Disorders/metabolism
- Growth Hormone/blood
- Growth Hormone/genetics
- Growth Hormone/metabolism
- Growth Hormone-Releasing Hormone/blood
- Growth Hormone-Releasing Hormone/genetics
- Growth Hormone-Releasing Hormone/pharmacology
- Insulin-Like Growth Factor Binding Protein 3/blood
- Insulin-Like Growth Factor Binding Protein 3/genetics
- Insulin-Like Growth Factor I/genetics
- Insulin-Like Growth Factor I/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutation
- Pituitary Gland/drug effects
- Pituitary Gland/metabolism
- Prolactin/genetics
- Prolactin/metabolism
- RGS Proteins/genetics
- RGS Proteins/metabolism
- Real-Time Polymerase Chain Reaction
- Receptors, Ghrelin/metabolism
- Receptors, Neuropeptide/genetics
- Receptors, Neuropeptide/metabolism
- Receptors, Pituitary Hormone-Regulating Hormone/genetics
- Receptors, Pituitary Hormone-Regulating Hormone/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Somatostatin/genetics
- Somatostatin/metabolism
- Somatostatin/pharmacology
Collapse
Affiliation(s)
- S Thierry Omouessi
- Department of Pharmacology, University of Michigan Medical School, Michigan, USA; Department of Physiology, Faculty of Medicine, Université des Sciences de la Santé (USS) de Libreville, Libreville, Gabon.
| | - Jeffrey R Leipprandt
- Department of Pharmacology and Toxicology, Michigan State University, Michigan, USA
| | - Marie-Yvonne Akoume
- International Research Institute of Biomedical Sciences & Biotechnology-Carles Kambangoye (IRBK), Université Internationale de Libreville, Essassa, Gabon; Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Sainte-Justine University Hospital Research Center, Montréal, Quebec, Canada
| | - Raelene Charbeneau
- Department of Pharmacology, University of Michigan Medical School, Michigan, USA
| | - Susan Wade
- Department of Pharmacology, University of Michigan Medical School, Michigan, USA
| | - Richard R Neubig
- Department of Pharmacology, University of Michigan Medical School, Michigan, USA; Department of Pharmacology and Toxicology, Michigan State University, Michigan, USA
| |
Collapse
|
13
|
Zhou W, Wang J, Zhang J, Peng C, Li G, Li D. Environmentally relevant concentrations of geosmin affect the development, oxidative stress, apoptosis and endocrine disruption of embryo-larval zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 735:139373. [PMID: 32473435 DOI: 10.1016/j.scitotenv.2020.139373] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 06/11/2023]
Abstract
Geosmin (trans-1, 10-dimethyl-trans-9-decalol), a volatile organic compound, has been widely detected in aquatic ecosystems. However, the ecological effects of geosmin are not clear. Here, using zebrafish (Danio rerio) embryo as a model, we investigated biological activity effects of environmentally relevant concentrations (50, 500, 5000 ng/L) of geosmin on the developing zebrafish starting from 2 h post-fertilization (hpf) to 96 hpf. Results showed geosmin had no effect on hatchability, malformations and mortality. However, we observed that geosmin exposure significantly increased zebrafish body length in a concentration dependent manner. This effect was possibly due to up-regulation of expression of genes along the growth hormone/insulin-like growth factor (GH/IGF) axis and hypothalamic-pituitary-thyroid (HPT) axis. In addition, superoxide dismutase (SOD) activities and catalase (CAT) activities significantly increased at 96 hpf when the embryos were exposed to 500 and 5000 ng/L of geosmin. The malondialdehyde (MDA) contents and glutathione S-transferase (GST) activities decreased significantly after the exposure to 5000 ng/L geosmin. Simultaneously, exposure to geosmin resulted in significant increase in cell apoptosis, mainly in the heart area. The mRNA levels of the genes related to oxidative stress and apoptosis were also altered significantly after geosmin exposure. These findings indicated that geosmin can simultaneously induce multiple responses during zebrafish embryonic development, including oxidative stress, apoptosis, and endocrine disruption.
Collapse
Affiliation(s)
- Weicheng Zhou
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; College of Chemistry, Biology and Environmental Engineering, Xiangnan University, Chenzhou 423000, PR China
| | - Jinglong Wang
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jinli Zhang
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Chengrong Peng
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, PR China
| | - Genbao Li
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, PR China
| | - Dunhai Li
- Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, PR China.
| |
Collapse
|
14
|
Cheng Y, Chen T, Song J, Teng Z, Wang C, Wang S, Lu G, Feng T, Qi Q, Xi Q, Liu S, Hao L, Zhang Y. Pituitary miRNAs target GHRHR splice variants to regulate GH synthesis by mediating different intracellular signalling pathways. RNA Biol 2020; 17:1754-1766. [PMID: 32508238 DOI: 10.1080/15476286.2020.1778295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Growth hormone (GH), whose synthesis and release are mainly regulated by intracellular signals mediated by growth hormone-releasing hormone receptor (GHRHR), is one of the major pituitary hormones and critical regulators of organism growth, metabolism, and immunoregulation. Pig GHRHR splice variants (SVs) may activate different signalling pathways via the variable C-terminal by alternative splicing, and SVs have the potential to change microRNA (miRNA) binding sites. In this study, we first confirmed the existence of pig GHRHR SVs (i.e., GHRHR, GHRHR SV1 and SV2) and demonstrated the inhibitory effects of critical pituitary miRNAs (i.e., let-7e and miR-328-5p) on GH synthesis and cell proliferation of primary pituitary cells. The SVs of GHRHR targeted by let-7e and miR-328-5p were predicted via bioinformatics analysis and verified by performing dual-luciferase reporter assays and detecting the expression of target transcripts. The differential responses of let-7e, and miR-328-5p to GH-releasing hormone and the changes in signalling pathways mediated by GHRHR suggested that let-7e and miR-328-5p were involved in GH synthesis mediated by GHRHR SVs, indicating that the two miRNAs played different roles by different ways. Finally, results showed that the protein coded by the GHRHR transcript regulated GH through the NO/NOS signalling pathway, whereas that coded by SV1 and SV2 regulated GH through the PKA/CREB signalling pathway, which was confirmed by the changes in signalling pathways after transfecting the expression vectors of GHRHR SVs to GH3 cells. To the best of our knowledge, this paper is the first to report pituitary miRNAs regulate GH synthesis by targeting the different SVs of GHRHR.
Collapse
Affiliation(s)
- Yunyun Cheng
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China.,Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University , Guangzhou, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University , Guangzhou, China
| | - Jie Song
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Zhaohui Teng
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China.,Research and Development Centre, Dalian Mogue Biotech Co., Ltd , Dalian, China
| | - Chunli Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Siyao Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Guanhong Lu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Tianqi Feng
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Qien Qi
- School of Life Science and Engineering, Foshan University , Foshan China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University , Guangzhou, China
| | - Songcai Liu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Linlin Hao
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University , Guangzhou, China
| |
Collapse
|
15
|
Cheng Y, Chen T, Song J, Qi Q, Wang C, Xi Q, Liu S, Hao L, Zhang Y. miR-709 inhibits GHRP6 induced GH synthesis by targeting PRKCA in pituitary. Mol Cell Endocrinol 2020; 506:110763. [PMID: 32084499 DOI: 10.1016/j.mce.2020.110763] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/02/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023]
Abstract
Pituitary growth hormone (GH) plays an essential role in processes of organism growth and metabolism. MicroRNA (miRNA) could also participate in diverse biological processes. However, the role of miRNA in the regulation of pituitary GH during the growth process remains unclear. In this study, we firstly confirmed that the second highly expressed pituitary miRNA (miR-709) significantly inhibited the GH synthesis and suppressed the viability of GH3 cells. The bioinformatics analysis and dual luciferase report system were used to ascertain the PRKCA is the direct target gene of miR-709, which is the coding gene of PKCα. Then the transcription and translation levels of Prkca were obvious reduced by the over-expression of miR-709 in GH3 cells, followed by the inhibition of the transcription factor (CREB1) of Gh1 gene and the ERK1/2 signaling pathway or the possible cross-talk signaling pathway (cAMP/PKA signaling pathway) detected by western blot, suggesting that ERK1/2 maybe an important factor involved in the GH3 cell viability mediated by PKCα. At last, GHRP6 increased PKCα and GH expression but reduced miR-709 expression in vitro and vivo assays, and this conclusion was further confirmed by the result of GHRP6 attenuated the inhibition of miR-709 on GH expression. These findings will provide new molecular mechanism on the regulation of pituitary GH.
Collapse
Affiliation(s)
- Yunyun Cheng
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510642, China
| | - Jie Song
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, 5333 Xian Road, Changchun, 130062, China
| | - Qien Qi
- School of Life Science and Engineering, Foshan University, Foshan, 528231, China
| | - Chunli Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, 5333 Xian Road, Changchun, 130062, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510642, China
| | - Songcai Liu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, 5333 Xian Road, Changchun, 130062, China
| | - Linlin Hao
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, 5333 Xian Road, Changchun, 130062, China.
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510642, China.
| |
Collapse
|
16
|
Li Z, Zhang N, Zhu L, Nan J, Shen J, Wang Z, Lin Y. Growth hormone-releasing hormone promotes therapeutic effects of peripheral blood endothelial progenitor cells in ischemic repair. J Endocrinol Invest 2020; 43:315-328. [PMID: 31506908 DOI: 10.1007/s40618-019-01109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 08/30/2019] [Indexed: 11/26/2022]
Abstract
PURPOSE In peripheral artery disease, blockage of the blood supply to the limbs leads to blood flow attenuation and tissue ischemia. We investigated whether growth hormone-releasing hormone (GHRH) could enhance the biological functions and therapeutic effects of endothelial progenitor cells (EPCs) derived from adult human peripheral blood (PB). METHODS EPCs were isolated from human PB (PB-EPCs) and cord blood and expanded in vitro. PB-EPCs incubated with or without GHRH were evaluated for proliferation, migration, and angiogenesis capacity and apoptosis rates under oxidative stress conditions. Activation of STAT3 and Akt pathways was evaluated using Western blot. A hind-limb ischemia (HLI) mouse model was used to study the efficacy of GHRH in improving EPC therapy in vivo. RESULTS GHRH enhanced the proliferation, migration, and angiogenesis capacity of PB-EPCs and reduced apoptosis under H2O2 stimulation. These beneficial effects were GHRH receptor-dependent and were paralleled by increased phosphorylation of STAT3 and Akt. Transplantation of GHRH-preconditioned EPCs into HLI model mice enhanced blood flow recovery by increasing vascular formation density and enhanced tissue regeneration at the lesion site. CONCLUSION Our studies demonstrate a novel role for GHRH in dramatically improving therapeutic angiogenesis in HLI by enhancing the biological functions of EPCs. These findings support additional studies to explore the full potential of GHRH in augmenting cell therapy for the management of ischemia.
Collapse
Affiliation(s)
- Z Li
- Research Institute of Experimental Neurobiology, Department of Neurology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - N Zhang
- Research Institute of Experimental Neurobiology, Department of Neurology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - L Zhu
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - J Nan
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - J Shen
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Z Wang
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Y Lin
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China.
| |
Collapse
|
17
|
Recinella L, Chiavaroli A, Orlando G, Ferrante C, Marconi GD, Gesmundo I, Granata R, Cai R, Sha W, Schally AV, Brunetti L, Leone S. Antinflammatory, antioxidant, and behavioral effects induced by administration of growth hormone-releasing hormone analogs in mice. Sci Rep 2020; 10:732. [PMID: 31959947 PMCID: PMC6971229 DOI: 10.1038/s41598-019-57292-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023] Open
Abstract
Growth hormone-releasing hormone (GHRH) antagonist MIA-690 and GHRH agonist MR-409, previously synthesized and developed by us have demonstrated potent antitumor effects. However, little is known about the effects of these analogs on brain functions. We investigated the potential antinflammatory and antioxidant effects of GHRH antagonist MIA-690 and GHRH agonist MR-409, on isolated mouse prefrontal cortex specimens treated with lipopolysaccharide (LPS). Additionally, we studied their effects on emotional behavior after chronic in vivo treatment. Ex vivo, MIA-690 and MR-409 inhibited LPS-induced inflammatory and pro-oxidative markers. In vivo, both MIA-690 and MR-409 induced anxiolytic and antidepressant-like effects, increased norepinephrine and serotonin levels and decreased nuclear factor-kB, tumor necrosis factor-α and interleukin-6 gene expression in prefrontal cortex. Increased nuclear factor erythroid 2–related factor 2 expression was also found in mice treated with MIA-690 and MR-409. MIA-690 showed higher efficacy in inhibiting all tested inflammatory and oxidative markers. In addition, MR-409 induced a down regulation of the gene and protein expression of pituitary-type GHRH-receptor in prefrontal cortex of mice after 4 weeks of treatment at 5 µg/day. In conclusion, our results demonstrate anxiolytic and antidepressant-like effects of GHRH analogs that could involve modulatory effects on monoaminergic signaling, inflammatory and oxidative status.
Collapse
Affiliation(s)
- Lucia Recinella
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy
| | | | | | | | | | - Iacopo Gesmundo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Riccarda Granata
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Renzhi Cai
- Veterans Affairs Medical Center, Miami, FL, 33125, USA.,Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.,Division of Medical/Oncology, Department of Pathology, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Wei Sha
- Veterans Affairs Medical Center, Miami, FL, 33125, USA.,Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.,Division of Medical/Oncology, Department of Pathology, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Andrew V Schally
- Veterans Affairs Medical Center, Miami, FL, 33125, USA.,Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.,Division of Medical/Oncology, Department of Pathology, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Luigi Brunetti
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy.
| | - Sheila Leone
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy.
| |
Collapse
|
18
|
Ford ZK, Dourson AJ, Liu X, Lu P, Green KJ, Hudgins RC, Jankowski MP. Systemic growth hormone deficiency causes mechanical and thermal hypersensitivity during early postnatal development. IBRO Rep 2019; 6:111-121. [PMID: 30815617 PMCID: PMC6378845 DOI: 10.1016/j.ibror.2019.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/06/2019] [Indexed: 12/22/2022] Open
Abstract
Systemic GHD causes behavioral hypersensitivity at P7 and P14, but not P21. Primary afferent sensitization is observed in GHRHr KOs. Knockout of GHRHr changes DRG gene expression that is observed throughout development.
Injury during early postnatal life causes acute alterations in afferent function and DRG gene expression, which in addition to producing short-term sensitivity has the potential to influence nociceptive responses in adulthood. We recently discovered that growth hormone (GH) is a key regulator of afferent sensitization and pain-related behaviors during developmental inflammation of the skin. Peripheral injury caused a significant reduction in cutaneous GH levels, which corresponded with the observed hypersensitivity. However, it has yet to be determined whether GH deficiency (GHD) is sufficient to drive peripheral sensitization in uninjured animals. Here, we found that systemic GHD, induced by knockout of the GH release hormone receptor (GHRHr), was able to induce behavioral and afferent hypersensitivity to peripheral stimuli specifically during early developmental stages. GHD also produced an upregulation of many receptors and channels linked to nociceptive processing in the DRGs at these early postnatal ages (P7 and P14). Surprisingly, P21 GHRHr knockouts also displayed significant alterations in DRG gene expression even though behavioral and afferent hypersensitivity resolved. These data support previous findings that GH is a key modulator of neonatal hypersensitivity. Results may provide insight into whether GH treatment may be a therapeutic strategy for pediatric pain.
Collapse
Affiliation(s)
- Zachary K. Ford
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, United States
| | - Adam J. Dourson
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, United States
| | - Xiaohua Liu
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, United States
| | - Peilin Lu
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, United States
| | - Kathryn J. Green
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, United States
| | - Renita C. Hudgins
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, United States
| | - Michael P. Jankowski
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, United States
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati OH 45229, United States
- Corresponding author at: Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave MLC 6016, Cincinnati, OH 45229, United States.
| |
Collapse
|
19
|
Dussor G, Boyd JT, Akopian AN. Pituitary Hormones and Orofacial Pain. Front Integr Neurosci 2018; 12:42. [PMID: 30356882 PMCID: PMC6190856 DOI: 10.3389/fnint.2018.00042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/06/2018] [Indexed: 12/15/2022] Open
Abstract
Clinical and basic research on regulation of pituitary hormones, extra-pituitary release of these hormones, distribution of their receptors and cell signaling pathways recruited upon receptor binding suggests that pituitary hormones can regulate mechanisms of nociceptive transmission in multiple orofacial pain conditions. Moreover, many pituitary hormones either regulate glands that produce gonadal hormones (GnH) or are regulated by GnH. This implies that pituitary hormones may be involved in sex-dependent mechanisms of orofacial pain and could help explain why certain orofacial pain conditions are more prevalent in women than men. Overall, regulation of nociception by pituitary hormones is a relatively new and emerging area of pain research. The aims of this review article are to: (1) present an overview of clinical conditions leading to orofacial pain that are associated with alterations of serum pituitary hormone levels; (2) discuss proposed mechanisms of how pituitary hormones could regulate nociceptive transmission; and (3) outline how pituitary hormones could regulate nociception in a sex-specific fashion. Pituitary hormones are routinely used for hormonal replacement therapy, while both receptor antagonists and agonists are used to manage certain pathological conditions related to hormonal imbalance. Administration of these hormones may also have a place in the treatment of pain, including orofacial pain. Hence, understanding the involvement of pituitary hormones in orofacial pain, especially sex-dependent aspects of such pain, is essential to both optimize current therapies as well as provide novel and sex-specific pharmacology for a diversity of associated conditions.
Collapse
Affiliation(s)
- Gregory Dussor
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, United States
| | - Jacob T Boyd
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Armen N Akopian
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Department of Pharmcology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
20
|
Sundralingam T, Tennekoon KH, de Silva S, De Silva S, Hewage S, Ranasinghe R. Novel gross deletion at the GHRHR gene locus possibly mediated by Alu specific microhomology identified in a Sri Lankan patient with isolated growth hormone deficiency. Growth Horm IGF Res 2018; 42-43:94-101. [PMID: 30390533 DOI: 10.1016/j.ghir.2018.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/14/2018] [Accepted: 10/20/2018] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Characterization of a deletion in the exon 1 and 5' regulatory region of the GHRHR gene in a proband with isolated growth hormone deficiency. METHODS Multiple ligation dependent probe amplification (MLPA) assay was carried out to confirm the homozygous deletion which was suspected during screening of the GHRHR gene by single strand conformation polymorphism. A series of short range PCR amplifications were carried out to map the approximate location of the break points of the deletion. Sanger sequencing was carried out to locate the break points and to identify the length of the deletion. Long range PCR amplification was carried out to confirm the length of the deletion and to screen the parents of the proband for the deletion. RESULTS A homozygous deletion was confirmed via MLPA assay. Zones of sequence similarity between upstream intergenic region and intron 1 of the GHRHR gene were identified. Break points of the deletion were identified within perfectly matching 32 bp repeat sequences ie: microhomologies in the specified zones. The novel deletion may have arisen via Alu specific microhomology mediated non-recurrent rearrangement in the maternal lineage of the proband. The deletion being reported in this study include, last 3118 bp from the upstream intergenic region and complete exon 1 and first 2620 bp from intron 1 and one of the 32 bp microhomologies. The total length of the deleted segment was 5875 bp. As the deleted region contained significant elements essential for gene expression, the identified deletion is being reported as likely pathogenic. The same deletion was identified in the mother in heterozygous state. CONCLUSION We have characterized a novel deletion that seems to have arisen via Alu specific microhomology mediated non-recurrent rearrangement at GHRHR gene locus. HGVS nomenclature of the deletion is c.-3166_58-2057del. This novel structural variant was identified to be the cause of IGHD of the affected proband.
Collapse
Affiliation(s)
- Tharmini Sundralingam
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, 90, Cumaratunga Munidasa Mawatha, Colombo 03, Sri Lanka
| | - Kamani Hemamala Tennekoon
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, 90, Cumaratunga Munidasa Mawatha, Colombo 03, Sri Lanka.
| | - Shamya de Silva
- Department of Paediatrics, Faculty of Medicine, University of Colombo, PO Box 271, Kynsey Road, Colombo 08, Sri Lanka; Lady Ridgeway Hospital, Dr. Danister de Silva Mawatha, Colombo 08, Sri Lanka
| | - Sumadee De Silva
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, 90, Cumaratunga Munidasa Mawatha, Colombo 03, Sri Lanka.
| | - Sudeshini Hewage
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, 90, Cumaratunga Munidasa Mawatha, Colombo 03, Sri Lanka.
| | - Ruwandi Ranasinghe
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, 90, Cumaratunga Munidasa Mawatha, Colombo 03, Sri Lanka.
| |
Collapse
|
21
|
Granata R. Peripheral activities of growth hormone-releasing hormone. J Endocrinol Invest 2016; 39:721-7. [PMID: 26891937 DOI: 10.1007/s40618-016-0440-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/03/2016] [Indexed: 12/21/2022]
Abstract
Growth hormone (GH)-releasing hormone (GHRH) is produced by the hypothalamus and stimulates GH synthesis and release in the anterior pituitary gland. In addition to its endocrine role, GHRH exerts a wide range of extrapituitary effects which include stimulation of cell proliferation, survival and differentiation, and inhibition of apoptosis. Accordingly, expression of GHRH, as well as the receptor GHRH-R and its splice variants, has been demonstrated in different peripheral tissues and cell types. Among the direct peripheral activities, GHRH regulates pancreatic islet and β-cell survival and function and endometrial cell proliferation, promotes cardioprotection and wound healing, influences the immune and reproductive systems, reduces inflammation, indirectly increases lifespan and adiposity and acts on skeletal muscle cells to inhibit cell death and atrophy. Therefore, it is becoming increasingly clear that GHRH exerts important extrapituitary functions, suggesting potential therapeutic use of the peptide and its analogs in a wide range of medical settings.
Collapse
Affiliation(s)
- R Granata
- Lab of Molecular and Cellular Endocrinology, Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Torino, Corso Dogliotti, 14, 10126, Turin, Italy.
| |
Collapse
|
22
|
Amyloid formation of growth hormone in presence of zinc: Relevance to its storage in secretory granules. Sci Rep 2016; 6:23370. [PMID: 27004850 PMCID: PMC4804206 DOI: 10.1038/srep23370] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/04/2016] [Indexed: 11/16/2022] Open
Abstract
Amyloids are cross-β-sheet fibrillar aggregates, associated with various human diseases and native functions such as protein/peptide hormone storage inside secretory granules of neuroendocrine cells. In the current study, using amyloid detecting agents, we show that growth hormone (GH) could be stored as amyloid in the pituitary of rat. Moreover, to demonstrate the formation of GH amyloid in vitro, we studied various conditions (solvents, glycosaminoglycans, salts and metal ions) and found that in presence of zinc metal ions (Zn(II)), GH formed short curvy fibrils. The amyloidogenic nature of these fibrils was examined by Thioflavin T binding, Congo Red binding, transmission electron microscopy and X-ray diffraction. Our biophysical studies also suggest that Zn(II) initiates the early oligomerization of GH that eventually facilitates the fibrillation process. Furthermore, using immunofluorescence study of pituitary tissue, we show that GH in pituitary significantly co-localizes with Zn(II), suggesting the probable role of zinc in GH aggregation within secretory granules. We also found that GH amyloid formed in vitro is capable of releasing monomers. The study will help to understand the possible mechanism of GH storage, its regulation and monomer release from the somatotrophs of anterior pituitary.
Collapse
|
23
|
Fridlyand LE, Tamarina NA, Schally AV, Philipson LH. Growth Hormone-Releasing Hormone in Diabetes. Front Endocrinol (Lausanne) 2016; 7:129. [PMID: 27777568 PMCID: PMC5056186 DOI: 10.3389/fendo.2016.00129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/06/2016] [Indexed: 12/13/2022] Open
Abstract
Growth hormone-releasing hormone (GHRH) is produced by the hypothalamus and stimulates growth hormone synthesis and release in the anterior pituitary gland. In addition, GHRH is an important regulator of cellular functions in many cells and organs. Expression of GHRH G-Protein Coupled Receptor (GHRHR) has been demonstrated in different peripheral tissues and cell types, including pancreatic islets. Among the peripheral activities, recent studies demonstrate a novel ability of GHRH analogs to increase and preserve insulin secretion by beta-cells in isolated pancreatic islets, which makes them potentially useful for diabetes treatment. This review considers the role of GHRHR in the beta-cell and addresses the unique engineered GHRH agonists and antagonists for treatment of type 2 diabetes mellitus. We discuss the similarity of signaling pathways activated by GHRHR in pituitary somatotrophs and in pancreatic beta-cells and possible ways as to how the GHRHR pathway can interact with glucose and other secretagogues to stimulate insulin secretion. We also consider the hypothesis that novel GHRHR agonists can improve glucose metabolism in Type 2 diabetes by preserving the function and survival of pancreatic beta-cells. Wound healing and cardioprotective action with new GHRH agonists suggest that they may prove useful in ameliorating certain diabetic complications. These findings highlight the future potential therapeutic effectiveness of modulators of GHRHR activity for the development of new therapeutic approaches in diabetes and its complications.
Collapse
Affiliation(s)
- Leonid E. Fridlyand
- Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
- *Correspondence: Leonid E. Fridlyand,
| | - Natalia A. Tamarina
- Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Andrew V. Schally
- VA Medical Center, Miami, FL, USA
- Department of Pathology and Medicine, Division of Endocrinology and Hematology-Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Louis H. Philipson
- Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
24
|
A hypothesis for a possible synergy between ghrelin and exercise in patients with cachexia: Biochemical and physiological bases. Med Hypotheses 2015; 85:927-33. [PMID: 26404870 DOI: 10.1016/j.mehy.2015.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 09/07/2015] [Accepted: 09/07/2015] [Indexed: 12/15/2022]
Abstract
This article reviews the biochemical and physiological observations underpinning the synergism between ghrelin and ghrelin agonists with exercise, especially progressive resistance training that has been shown to increase muscle mass. The synergy of ghrelin agonists and physical exercise could be beneficial in conditions where muscle wasting is present, such as that found in patients with advanced cancer. The principal mechanism that controls muscle anabolism following the activation of the ghrelin receptor in the central nervous system involves the release of growth hormone/insulin-like growth factor-1 (GH/IGF-1). GH/IGF-1 axis has a dual pathway of action on muscle growth: (a) a direct action on muscle, bone and fat tissue and (b) an indirect action via the production of both muscle-restricted mIGF-1 and anti-cachectic cytokines. Progressive resistance training is a potent inducer of the secretion the muscle-restricted IGF-1 (mIGF-1) that enhances protein synthesis, increases lean body mass and eventually leads to the improvement of muscle strength. Thus, the combination of ghrelin administration with progressive resistance training may serve to circumvent ghrelin resistance and further reduce muscle wasting, which are commonly associated with cachexia.
Collapse
|
25
|
Qi QE, Xi QY, Ye RS, Chen T, Cheng X, Li CY, Zhu XT, Shu G, Wang LN, Jiang QY, Zhang YL. Alteration of the miRNA expression profile in male porcine anterior pituitary cells in response to GHRH and CST and analysis of the potential roles for miRNAs in regulating GH. Growth Horm IGF Res 2015; 25:66-74. [PMID: 25613666 DOI: 10.1016/j.ghir.2014.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 11/27/2014] [Accepted: 12/01/2014] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Growth hormone releasing hormone (GHRH) is a major positive regulator of growth hormone (GH) in the anterior pituitary gland, while cortistatin's (CST) role is negative. miRNAs (microRNAs or miRs) are small RNA molecules modulating gene expression at the post-transcriptional level. However, little is known about the function of miRNAs in the regulation of GH synthesis and/or secretion. This study investigated potential functional miRNAs involved in GH secretion in the normal porcine pituitary. DESIGN Primary porcine anterior pituitary cells were cultivated and then treated with 10 nmol/L GHRH and 100 nmol/L CST, respectively. The effects of GHRH and CST on GH secretion were determined using RIA. miRNA microarrays were employed to analyze miRNA expression after treatment and then differentially expressed miRNAs were screened. Bioinformatics analysis was used to analyze the potential targets in growth hormone regulation of altered miRNAs. Furthermore, functional experiments were conducted to study the function of ssc-let-7c. RESULTS GHRH significantly promoted GH secretion, while CST suppressed GH secretion. 19 and 35 differentially expressed miRNAs were identified in response to GHRH and CST treatments respectively. Verification of 5 randomly selected miRNAs by quantitative real-time PCR (qRT-PCR) showed similar changes with microarray analysis. Target analysis showed that some miRNAs may be involved in GH secretion-related pathways. Importantly, ssc-let-7c was predicted to target GH1 and GHRHR mRNA 3'untranslated regions (3'UTRs), which was supported by luciferase reporter assay. Furthermore, functional experimental results showed that ssc-let-7c was involved in GH secretion regulation, and overexpression of ssc-let-7c inhibited GH secretion in porcine anterior pituitary cells. CONCLUSIONS GHRH and CST modulated porcine pituitary cell miRNA expression. Bioinformatics analysis revealed a complicated network among differentially expressed miRNAs, GH regulation-related genes and hormones. More interestingly, ssc-let-7c inhibited both GH1 and GHRHR mRNA 3'UTR reporter vectors' luciferase activity and overexpression of ssc-let-7c led to a decrease of GH secretion.
Collapse
Affiliation(s)
- Qi-En Qi
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China
| | - Qian-Yun Xi
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China
| | - Rui-Song Ye
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China
| | - Ting Chen
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China
| | - Xiao Cheng
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China
| | - Chao-Yun Li
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China
| | - Xiao-Tong Zhu
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China
| | - Gang Shu
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China
| | - Li-Na Wang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China
| | - Qing-Yan Jiang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China
| | - Yong-Liang Zhang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Wushan Road, Guangzhou 510642, China.
| |
Collapse
|
26
|
Antagonist of GH-releasing hormone receptors alleviates experimental ocular inflammation. Proc Natl Acad Sci U S A 2014; 111:18303-8. [PMID: 25489106 DOI: 10.1073/pnas.1421815112] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Disruptions in immunity and occurrence of inflammation cause many eye diseases. The growth hormone-releasing hormone-growth hormone-insulin-like growth factor-1 (GHRH-GH-IGF1) axis exerts regulatory effects on the immune system. Its involvement in ocular inflammation remains to be investigated. Here we studied this signaling in endotoxin-induced uveitis (EIU) generated by LPS. The increase in GHRH receptor (GHRH-R) protein levels was parallel to the increase in mRNA levels of pituitary-specific transcription factor-1, GHRH-R splice variant 1, GHRH, and GH following LPS insult. Elevation of GHRH-R and GH receptor was localized on the epithelium of the iris and ciliary body, and GHRH-R was confined to the infiltrating macrophages and leukocytes in aqueous humor but not to those in stroma. Treatment with GHRH-R antagonist decreased LPS-stimulated surges of GH and IGF1 in aqueous humor and alleviated inflammation by reducing the infiltration of macrophages and leukocytes and the production of TNF-α, IL-1β, and monocyte chemotactic protein-1. Our results indicate that inflammation in the iris and ciliary body involves the activation of GHRH signaling, which affects the recruitment of immune cells and the production of proinflammatory mediators that contribute to EIU pathogenesis. Moreover, the results suggest that GHRH-R antagonists are potential therapeutic agents for the treatment of acute ocular inflammation.
Collapse
|
27
|
Arman A, Dündar BN, Çetinkaya E, Erzaim N, Büyükgebiz A. Novel growth hormone-releasing hormone receptor gene mutations in Turkish children with isolated growth hormone deficiency. J Clin Res Pediatr Endocrinol 2014; 6:202-8. [PMID: 25541890 PMCID: PMC4293654 DOI: 10.4274/jcrpe.1518] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE Isolated growth hormone deficiency (IGHD) is defined as a medical condition associated with growth failure due to insufficient production of GH or lack of GH action. Mutations in the gene encoding for GH-releasing hormone receptor (GHRHR) have been detected in patients with IGHD type IB. However, genetic defects on GHRHR causing IGHD in the Turkish population have not yet been reported. To identify mutations on GHRHR gene in a population of Turkish children with IGHD. METHODS Ninety-six Turkish children with IGHD were included in this study. Exon1-13 and exon/intron boundaries of GHRHR were amplified by suitable primers. The polymerase chain reaction products for GHRHR gene were sequenced with primers. RESULTS We analyzed the GHRHR gene for mutations in ninety-six patients with IGHD based on sequence results. We identified novel p.K264E, p.S317T, p.S330L, p.G369V, p.T257A and C base insertion on position 380 (c.380inserC) mutations. In 5 of the patients, the mutation was homozygote and in 1-heterozygote (p.S317T). CONCLUSION Six new missense mutations and one first case of insertion mutations for the GHRHR gene are reported.
Collapse
Affiliation(s)
- Ahmet Arman
- Marmara University Faculty of Medicine, Department of Medical Genetics, İstanbul, Turkey. E-ma-il:
| | - Bumin Nuri Dündar
- Katip Çelebi University Faculty of Medicine, Department of Pediatric Endocrinology, İzmir, Turkey
| | | | - Nilüfer Erzaim
- Yeditepe University Faculty of Medicine, Department of Genetics and Bioengineering, İstanbul, Turkey
| | - Atilla Büyükgebiz
- Bilim University Faculty of Medicine, Department of Pediatric Endocrinology, İstanbul, Turkey
| |
Collapse
|
28
|
Kale S, Budyal S, Kasaliwal R, Shivane V, Raghavan V, Lila A, Bandgar T, Shah N. A novel gross indel in the growth hormone releasing hormone receptor gene of Indian IGHD patients. Growth Horm IGF Res 2014; 24:227-232. [PMID: 25153028 DOI: 10.1016/j.ghir.2014.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 07/29/2014] [Indexed: 11/20/2022]
Abstract
CONTEXT Cohort specific mutations in the growth hormone (GH1) and growth hormone-releasing hormone receptor (GHRHR) genes have been reported worldwide in isolated growth hormone deficiency (IGHD) patients. However, limited data is available on ethnically diverse Indian IGHD patients. OBJECTIVE The aim of the study was to find GH1 and GHRHR gene mutations in Indian IGHD patients from two unrelated non-consanguineous families. DESIGN The 5' and 3' untranslated regions (UTRs) and coding regions with splice sites of the GH1 and GHRHR genes were sequenced for all patients (n=6). Family members and 20 controls were evaluated for the sequence variants identified in the index patients. Online bioinformatics tools were used to confirm mutations and their pathogenicity. RESULTS GHRHR gene mutations were observed in all patients. Interestingly, a novel indel g.30999250_31006943delinsAGAGATCCA was observed in both the unrelated families. Three patients were homozygous for the novel indel, two were homozygous for the previously reported p.E72X mutation and one was compound heterozygous with both the mutations (indel and p.E72X) in the GHRHR gene. The novel indel has resulted in the loss of 5' regulatory region and exon 1 of the GHRHR gene impairing the GHRHR expression. All the normal family members were heterozygous either for the indel or p.E72X mutation. None of the patients had GH1 gene mutations. CONCLUSIONS We describe a novel gross indel in the GHRHR gene resulting in the loss of 5' regulatory region and GHRHR exon 1 in four IGHD IB patients from two unrelated non-consanguineous Indian families.
Collapse
Affiliation(s)
- Shantanu Kale
- Department of Endocrinology, Seth G.S. Medical College, K.E.M. Hospital, Parel, Mumbai 400012, India.
| | - Sweta Budyal
- Department of Endocrinology, Seth G.S. Medical College, K.E.M. Hospital, Parel, Mumbai 400012, India
| | - Rajeev Kasaliwal
- Department of Endocrinology, Seth G.S. Medical College, K.E.M. Hospital, Parel, Mumbai 400012, India
| | - Vyankatesh Shivane
- Department of Endocrinology, Seth G.S. Medical College, K.E.M. Hospital, Parel, Mumbai 400012, India
| | - Vijaya Raghavan
- Department of Endocrinology, Seth G.S. Medical College, K.E.M. Hospital, Parel, Mumbai 400012, India
| | - Anurag Lila
- Department of Endocrinology, Seth G.S. Medical College, K.E.M. Hospital, Parel, Mumbai 400012, India
| | - Tushar Bandgar
- Department of Endocrinology, Seth G.S. Medical College, K.E.M. Hospital, Parel, Mumbai 400012, India
| | - Nalini Shah
- Department of Endocrinology, Seth G.S. Medical College, K.E.M. Hospital, Parel, Mumbai 400012, India
| |
Collapse
|
29
|
Agonists of growth hormone-releasing hormone stimulate self-renewal of cardiac stem cells and promote their survival. Proc Natl Acad Sci U S A 2014; 111:17260-5. [PMID: 25404316 DOI: 10.1073/pnas.1420375111] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The beneficial effects of agonists of growth hormone-releasing hormone receptor (GHRH-R) in heart failure models are associated with an increase in the number of ckit(+) cardiac stem cells (CSCs). The goal of the present study was to determine the presence of GHRH-R in CSCs, the effect of GHRH-R agonists on their proliferation and survival, and the mechanisms involved. We investigated the expression of GHRH-R in CSCs of different species and the effect of GHRH-R agonists on their cell proliferation and survival. GHRH-R is expressed in ckit(+) CSCs isolated from mouse, rat, and pig. Treatment of porcine CSCs with the GHRH-R agonist JI-38 significantly increased the rate of cell division. Similar results were observed with other GHRH-R agonists, MR-356 and MR-409. JI-38 exerted a protective effect on survival of porcine CSCs under conditions of oxidative stress induced by exposure to hydrogen peroxide. Treatment with JI-38 before exposure to peroxide significantly reduced cell death. A similar effect was observed with MR-356. Addition of GHRH-R agonists to porcine CSCs induced activation of ERK and AKT pathways as determined by increased expression of phospho-ERK and phospho-AKT. Inhibitors of ERK and AKT pathways completely reversed the effect of GHRH-R agonists on CSC proliferation. Our findings extend the observations of the expression of GHRH-R by CSCs and demonstrate that GHRH-R agonists have a direct effect on proliferation and survival of CSCs. These results support the therapeutic use of GHRH-R agonists for stimulating endogenous mechanisms for myocardial repair or for preconditioning of stem cells before transplantation.
Collapse
|
30
|
Cai R, Schally AV, Cui T, Szalontay L, Halmos G, Sha W, Kovacs M, Jaszberenyi M, He J, Rick FG, Popovics P, Kanashiro-Takeuchi R, Hare JM, Block NL, Zarandi M. Synthesis of new potent agonistic analogs of growth hormone-releasing hormone (GHRH) and evaluation of their endocrine and cardiac activities. Peptides 2014; 52:104-12. [PMID: 24373935 PMCID: PMC4745889 DOI: 10.1016/j.peptides.2013.12.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 12/17/2013] [Accepted: 12/18/2013] [Indexed: 11/30/2022]
Abstract
In view of the recent findings of stimulatory effects of GHRH analogs, JI-34, JI-36 and JI-38, on cardiomyocytes, pancreatic islets and wound healing, three series of new analogs of GHRH(1-29) have been synthesized and evaluated biologically in an endeavor to produce more potent compounds. "Agmatine analogs", MR-356 (N-Me-Tyr(1)-JI-38), MR-361(N-Me-Tyr(1), D-Ala(2)-JI-38) and MR-367(N-Me-Tyr(1), D-Ala(2), Asn(8)-JI-38), in which Dat in JI-38 is replaced by N-Me-Tyr(1), showed improved relative potencies on GH release upon subcutaneous administration in vivo and binding in vitro. Modification with N-Me-Tyr(1) and Arg(29)-NHCH3 as in MR-403 (N-Me-Tyr(1), D-Ala(2), Arg(29)-NHCH3-JI-38), MR-406 (N-Me-Tyr(1), Arg(29)-NHCH3-JI-38) and MR-409 (N-Me-Tyr(1), D-Ala(2), Asn(8), Arg(29)-NHCH3-JI-38), and MR-410 (N-Me-Tyr(1), D-Ala(2), Thr(8), Arg(29)-NHCH3-JI-38) resulted in dramatically increased endocrine activities. These appear to be the most potent GHRH agonistic analogs so far developed. Analogs with Apa(30)-NH2 such as MR-326 (N-Me-Tyr(1), D-Ala(2), Arg(29), Apa(30)-NH2-JI-38), and with Gab(30)-NH2, as MR-502 (D-Ala(2), 5F-Phe(6), Ser(28), Arg(29),Gab(30)-NH2-JI-38) also exhibited much higher potency than JI-38 upon i.v. administration. The relationship between the GH-releasing potency and the analog structure is discussed. Fourteen GHRH agonists with the highest endocrine potencies were subjected to cardiologic tests. MR-409 and MR-356 exhibited higher potency than JI-38 in activating myocardial repair in rats with induced myocardial infarction. As the previous class of analogs, exemplified by JI-38, had shown promising results in multiple fields including cardiology, diabetes and wound healing, our new, more potent, GHRH agonists should manifest additional efficacy for possible medical applications.
Collapse
Affiliation(s)
- Renzhi Cai
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Andrew V Schally
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, United States; Division of Hematology/Oncology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, United States; Division of Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, United States.
| | - Tengjiao Cui
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Luca Szalontay
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States
| | - Gabor Halmos
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Wei Sha
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States; Interdisciplinary Stem Cell Institute, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Magdolna Kovacs
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Miklos Jaszberenyi
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States
| | - Jinlin He
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States
| | - Ferenc G Rick
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States; Department of Urology, Florida International University, Herbert Wertheim College of Medicine, Miami, FL, United States
| | - Petra Popovics
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; Division of Cardiology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Rosemeire Kanashiro-Takeuchi
- Interdisciplinary Stem Cell Institute, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Joshua M Hare
- Division of Cardiology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, United States; Interdisciplinary Stem Cell Institute, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Norman L Block
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, United States; Division of Hematology/Oncology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Marta Zarandi
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States; South Florida VA Foundation for Research and Education, Miami, FL, United States; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
31
|
A 5'UTR SNP of GHRHR locus is associated with body weight and average daily gain in Chinese cattle. Mol Biol Rep 2012; 39:10469-73. [PMID: 23053950 DOI: 10.1007/s11033-012-1927-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 10/01/2012] [Indexed: 10/27/2022]
Abstract
Growth hormone-releasing hormone receptor (GHRHR) has important functions in the regulation of the growth hormone axis and the development and proliferation of pituitary somatotropes. Moreover, some mutations in mouse GHRHR can induce the dwarfism. The objective of this paper is to reveal the association of GHRHR with growth traits in three Chinese cattle breeds, including Nanyang cattle (NY, 220), Qinchuan cattle (QC, 114), and Jiaxian cattle (JX, 142). A novel single nucleotide polymorphism (NM_181020:c.102C>T) in 5'UTR of GHRHR was identified using PCR-SSCP and DNA sequencing. The frequency of NM_181020:c.102C allele ranged from 0.926 to 0.956. We found that the locus was significantly associated with NY cattle's body weight (BW) of 6 months, with average daily gain (ADG) of 0-6 months, and as well as with ADG of 6-12 months (p < 0.05). The data suggested that the polymorphism (NM_181020:c.102C>T) of the GHRHR could be a molecular marker candidate for breeding of NY cattle in favor of BW.
Collapse
|
32
|
Liu W, Yu Y, Li G, Tang S, Zhang Y, Wang Y, Zhang S, Zhang Y. Single-nucleotide polymorphisms in the promoter of the growth hormone-releasing hormone receptor gene are associated with growth and reproduction traits in chickens. Anim Genet 2012; 43:564-9. [PMID: 22497307 DOI: 10.1111/j.1365-2052.2011.02306.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2011] [Indexed: 11/28/2022]
Abstract
Growth hormone-releasing hormone receptor (GHRHR) plays a critical role in growth hormone (GH) synthesis, release and regulation in animals. The objective of this study was to investigate variations of the chicken GHRHR gene and their associations with growth and reproduction traits in 768 Beijing You chickens. Results revealed three single nucleotide polymorphisms (SNPs) in the promoter region of the gene (g.-1654A>G, g.-1411A>G and g.-142T>C). Association analysis revealed that the novel SNP g.-1654A>G had significant effects on chicken body weight at 7, 9, 11, 13, 17 weeks of age and the age of first egg as well as egg number at 32, 36 and 40 weeks. Significant association was also observed between g.-1411A>G and g.-142T>C with EN24. Moreover, the age of first egg was distinctly related with g.-142T>C (P < 0.05). Although significant statistical difference was not detected in GHRHR mRNA levels among genotypes of the SNPs (P > 0.05), strong expression variations of the gene were found between the ages 17 and 20 weeks in the population (P < 0.05). These results suggest that the three SNPs in the GHRHR promoter could be used as potential genetic markers to improve the growth and reproductive traits in chickens.
Collapse
Affiliation(s)
- W Liu
- Key Laboratory of Agricultural Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Komatsu M, Kojima M, Okamura H, Nishio M, Kaneda M, Kojima T, Takeda H, Malau-Aduli AEO, Takahashi H. Age-related changes in gene expression of the growth hormone secretagogue and growth hormone-releasing hormone receptors in Holstein-Friesian cattle. Domest Anim Endocrinol 2012; 42:83-93. [PMID: 22056236 DOI: 10.1016/j.domaniend.2011.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Revised: 09/23/2011] [Accepted: 09/26/2011] [Indexed: 10/15/2022]
Abstract
Growth hormone secretion from the anterior pituitary gland is controlled by interactions between three hormone receptors, between GHRH and GHRH receptor (GHRH-R), between ghrelin and growth hormone secretagogue receptor (GHS-R1a), and between somatostatin and somatostatin receptors in the hypothalamus and anterior pituitary gland. Ghrelin-GHS-R1a is involved in many important functions, including GH secretion and appetite. We investigated age-related changes in the expressions of GHS-R1a, GHS-R1b (the truncated-type receptor), and GHRH-R mRNAs by real-time reverse transcription-PCR using 16 tissues, leukocytes, oocytes, and cumulus cells in Holstein-Friesian cattle. The tissue samples were divided into three age classes: 1) 19 to 26 d of age (preweaning calves), 2) 2 mo to 6.5 mo of age (postweaning calves), and 3) 3.2 to 8.1 yr of age (cows). The GHS-R1a mRNA was highly (P < 0.05) expressed in the arcuate nucleus, pituitary gland, and liver compared with that of the other tissues in all age classes. Expression of GHS-R 1a mRNA in the arcuate nucleus of postweaning calves was > 10-fold greater (P < 0.01) than those of preweaning calves and cows, and its expression level was the greatest (P < 0.01) in all tissues examined in age group 2. GHS-R1a and GHRH-R mRNA expressions in the pituitary gland of preweaning calves tended to be greater (P < 0.20 and P < 0.17, respectively) than those of postweaning calves and cows. GHS-R1b mRNA expression was detected in all tissues examined, and abundance was greater (P < 0.05) in the pancreas, pituitary gland, spleen, arcuate nucleus, adipose tissue, and leukocyte compared with that of the other tissues examined in age group 3. Interestingly, a relatively large animal-to-animal variation was observed in pancreas GHS-R 1b mRNA expression. The GHRH-R mRNA was markedly increased (P < 0.01) in the pituitary gland in all age groups compared with that of the other tissues. GHRH-R mRNA abundance in the arcuate nucleus, pituitary gland, liver, spleen, adipose tissue, and heart of preweaning calves tended to be greater than those of postweaning calves and cows. The GHRH-R mRNA was not detected in the mammary gland and adipose tissue of nonlactating cows.
Collapse
MESH Headings
- Age Factors
- Animals
- Arcuate Nucleus of Hypothalamus/physiology
- Cattle/genetics
- Cattle/metabolism
- Female
- Gene Expression Regulation
- Growth Hormone/biosynthesis
- Growth Hormone/genetics
- Growth Hormone/metabolism
- Least-Squares Analysis
- Male
- Pituitary Gland, Anterior/metabolism
- Pituitary Gland, Anterior/physiology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction/veterinary
- Receptors, Ghrelin/biosynthesis
- Receptors, Ghrelin/genetics
- Receptors, Neuropeptide/biosynthesis
- Receptors, Neuropeptide/genetics
- Receptors, Pituitary Hormone-Regulating Hormone/biosynthesis
- Receptors, Pituitary Hormone-Regulating Hormone/genetics
Collapse
Affiliation(s)
- M Komatsu
- National Institute of Livestock and Grassland Science, Ikenodai 2, Tsukuba, Ibaraki 305-0901, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Córdoba-Chacón J, Gahete MD, Castaño JP, Kineman RD, Luque RM. Homologous and heterologous in vitro regulation of pituitary receptors for somatostatin, growth hormone (GH)-releasing hormone, and ghrelin in a nonhuman primate (Papio anubis). Endocrinology 2012; 153:264-72. [PMID: 22109886 PMCID: PMC3249678 DOI: 10.1210/en.2011-1677] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Secretion of GH by pituitary somatotrophs is primarily stimulated by GHRH and ghrelin and inhibited by somatostatin through the activation of specific receptors [GHRH receptor (GHRH-R), GH secretagogue receptor (GHS-R) and somatostatin receptors (sst1-5), respectively]. However, we have shown that somatostatin, at low doses, can also stimulate GH release, directly and specifically, in primary pituitary cultures from a nonhuman primate (baboons, Papio anubis) and pigs. To determine whether somatostatin, GHRH, and ghrelin can also regulate the expression of their receptors in primates, pituitary cultures from baboons were treated for 4 h with GHRH or ghrelin (10(-8) m) or with high (10(-7) m) and low (10(-15) m) doses of somatostatin, and GH release and expression levels of all receptors were measured. GHRH/ghrelin decreased the expression of their respective receptors (GHRH-R and GHS-R). Both peptides increased sst1, only GHRH decreased sst5 expression, whereas sst2 expression remained unchanged. The effects of GHRH/ghrelin were completely mimicked by forskolin (adenylate cyclase activator) and phorbol 12-myristate 13-acetate (protein kinase C activator), respectively, indicating the regulation of receptor subtype levels by GHRH and ghrelin involved distinct signaling pathways. In contrast, high-dose somatostatin did not alter GH release but increased sst1, sst2, and sst5 expression, whereas GHRH-R and GHS-R expression were unaffected. Interestingly, low-dose somatostatin increased GH release and sst1 mRNA but decreased sst5 and GHRH-R expression, similar to that observed for GHRH. Altogether, our data show for the first time in a primate model that the primary regulators of somatotroph function (GHRH/ghrelin/somatostatin) exert both homologous and heterologous regulation of receptor synthesis which is dose and subtype dependent and involves distinct signaling pathways.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Colforsin/pharmacology
- Gene Expression/drug effects
- Ghrelin/administration & dosage
- Growth Hormone-Releasing Hormone/administration & dosage
- In Vitro Techniques
- Papio anubis/genetics
- Papio anubis/metabolism
- Pituitary Gland/drug effects
- Pituitary Gland/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Ghrelin/genetics
- Receptors, Ghrelin/metabolism
- Receptors, LHRH/genetics
- Receptors, LHRH/metabolism
- Receptors, Somatostatin/agonists
- Receptors, Somatostatin/genetics
- Receptors, Somatostatin/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Somatostatin/administration & dosage
- Swine
- Tetradecanoylphorbol Acetate/pharmacology
Collapse
Affiliation(s)
- Jose Córdoba-Chacón
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, E-14014 Córdoba, Spain
| | | | | | | | | |
Collapse
|
35
|
Activation of growth hormone releasing hormone (GHRH) receptor stimulates cardiac reverse remodeling after myocardial infarction (MI). Proc Natl Acad Sci U S A 2011; 109:559-63. [PMID: 22203988 DOI: 10.1073/pnas.1119203109] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Both cardiac myocytes and cardiac stem cells (CSCs) express the receptor of growth hormone releasing hormone (GHRH), activation of which improves injury responses after myocardial infarction (MI). Here we show that a GHRH-agonist (GHRH-A; JI-38) reverses ventricular remodeling and enhances functional recovery in the setting of chronic MI. This response is mediated entirely by activation of GHRH receptor (GHRHR), as demonstrated by the use of a highly selective GHRH antagonist (MIA-602). One month after MI, animals were randomly assigned to receive: placebo, GHRH-A (JI-38), rat recombinant GH, MIA-602, or a combination of GHRH-A and MIA-602, for a 4-wk period. We assessed cardiac performance and hemodynamics by using echocardiography and micromanometry derived pressure-volume loops. Morphometric measurements were carried out to determine MI size and capillary density, and the expression of GHRHR was assessed by immunofluorescence and quantitative RT-PCR. GHRH-A markedly improved cardiac function as shown by echocardiographic and hemodynamic parameters. MI size was substantially reduced, whereas myocyte and nonmyocyte mitosis was markedly increased by GHRH-A. These effects occurred without increases in circulating levels of growth hormone and insulin-like growth factor I and were, at least partially, nullified by GHRH antagonism, confirming a receptor-mediated mechanism. GHRH-A stimulated CSCs proliferation ex vivo, in a manner offset by MIA-602. Collectively, our findings reveal the importance of the GHRH signaling pathway within the heart. Therapy with GHRH-A although initiated 1 mo after MI substantially improved cardiac performance and reduced infarct size, suggesting a regenerative process. Therefore, activation of GHRHR provides a unique therapeutic approach to reverse remodeling after MI.
Collapse
|
36
|
Granata R, Isgaard J, Alloatti G, Ghigo E. Cardiovascular actions of the ghrelin gene-derived peptides and growth hormone-releasing hormone. Exp Biol Med (Maywood) 2011; 236:505-514. [DOI: 10.1258/ebm.2011.010365] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
In 1976, small peptide growth hormone secretagogues (GHSs) were discovered and found to promote growth hormone (GH) release from the pituitary. The GHS receptor (GHS-R) was subsequently cloned, and its endogenous ligand ghrelin was later isolated from the stomach. Ghrelin is a 28-amino acid peptide, whose acylation is essential for binding to GHS-R type 1a and for the endocrine functions, including stimulation of GH secretion and subsequent food intake. Unacylated ghrelin, the other ghrelin form, although devoid of GHS-R binding is an active peptide, sharing many peripheral effects with acylated ghrelin (AG). The ghrelin system is broadly expressed in myocardial tissues, where it exerts different functions. Indeed, ghrelin inhibits cardiomyocyte and endothelial cell apoptosis, and improves left ventricular (LV) function during ischemia–reperfusion (I/R) injury. In rats with heart failure (HF), ghrelin improves LV dysfunction and attenuates the development of cardiac cachexia. Similarly, ghrelin exerts vasodilatory effects in humans, improves cardiac function and decreases systemic vascular resistance in patients with chronic HF. Obestatin is a recently identified ghrelin gene peptide. The physiological role of obestatin and its binding to the putative GPR39 receptor are still unclear, although protective effects have been demonstrated in the pancreas and heart. Similarly to AG, the hypothalamic peptide growth hormone-releasing hormone (GHRH) stimulates GH release from the pituitary, through binding to the GHRH-receptor. Besides its proliferative effects in different cell types, at the cardiovascular level GHRH inhibits cardiomyocyte apoptosis, and reduces infarct size in both isolated rat heart after I/R and in vivo after myocardial infarction. Therefore, both ghrelin and GHRH exert cardioprotective effects, which make them candidate targets for therapeutic intervention in cardiovascular dysfunctions.
Collapse
Affiliation(s)
- Riccarda Granata
- Laboratory of Molecular and Cellular Endocrinology, Division of Endocrinology, Diabetology and Metabolism, Department of Internal Medicine, University of Turin, Corso Dogliotti, 14-10126 Turin, Italy
| | - Jörgen Isgaard
- Department of Internal Medicine, The Sahlgrenska Academy at the University of Gothenburg, 413 46 Gothenburg, Sweden
| | - Giuseppe Alloatti
- Department of Animal and Human Biology, University of Turin, 10123 Turin, Italy
| | - Ezio Ghigo
- Laboratory of Molecular and Cellular Endocrinology, Division of Endocrinology, Diabetology and Metabolism, Department of Internal Medicine, University of Turin, Corso Dogliotti, 14-10126 Turin, Italy
| |
Collapse
|
37
|
Zhang B, Zhao G, Lan X, Lei C, Zhang C, Chen H. Polymorphism in GHRH gene and its association with growth traits in Chinese native cattle. Res Vet Sci 2011; 92:243-6. [PMID: 21353268 DOI: 10.1016/j.rvsc.2011.01.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2010] [Revised: 01/13/2011] [Accepted: 01/23/2011] [Indexed: 11/29/2022]
Abstract
Growth hormone-releasing hormone (GHRH) is secreted by the hypothalamus and stimulates growth hormone (GH) released from the pituitary. Mutations detected in GHRH gene showed associations with animal production traits. The purpose of this study was to investigate the association of the GHRH gene with growth traits in Chinese native cattle. PCR-SSCP and sequencing were used to detect mutations of the GHRH gene in this study. One novel mutation 4251nt (C>T) was found and the frequencies of C allele were 0.8778 and 0.8476 for Qinchuan and Nanyang cattle, respectively. Body weight with the CT genotype was significantly higher (P<0.05 or P<0.01) than those with CC genotype for different growth periods (6, 12, 18, and 24 months old) in Nanyang cattle. Our findings suggested that polymorphism in bovine GHRH might be one of the important genetic factors to influence body weight.
Collapse
Affiliation(s)
- Bao Zhang
- College of Animal Science and Technology, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, China
| | | | | | | | | | | |
Collapse
|
38
|
Inoue H, Kangawa N, Kinouchi A, Sakamoto Y, Kimura C, Horikawa R, Shigematsu Y, Itakura M, Ogata T, Fujieda K. Identification and functional analysis of novel human growth hormone-releasing hormone receptor (GHRHR) gene mutations in Japanese subjects with short stature. Clin Endocrinol (Oxf) 2011; 74:223-33. [PMID: 21044116 DOI: 10.1111/j.1365-2265.2010.03911.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
CONTEXT Growth hormone-releasing hormone receptor (GHRHR) gene mutations have been identified in patients of different ethnic origins with isolated GH deficiency (IGHD) type IB. However, the prevalence of these mutations in the Japanese population has yet to be fully determined. OBJECTIVES This study aimed to evaluate the contributions of GHRHR mutations to the molecular mechanism underlying short stature in Japanese subjects. DESIGN The GHRHR gene was sequenced in 127 unrelated Japanese patients with either IGHD (n = 14) or idiopathic short stature (ISS; n = 113). Sequence variants were evaluated in family members and 188 controls, and then examined in functional studies. RESULTS A novel homozygous E382E (c.1146G>A) synonymous variant, at the last base of exon 12, was identified in an IGHD family with two affected sisters. In vitro splicing studies showed this mutation to result in skipping of exon 12. In one ISS patient, a heterozygous ATG-166T>C variant was found in the distal Pit-1 P2 binding element of the GHRHR promoter. In two control subjects, a close but distinct variant, ATG-164T>C, was detected. Functional studies showed that both promoter variants diminish promoter activity by altering Pit-1 binding ability. Four missense variants were also found in both patient and control groups but had no detectable functional consequences. CONCLUSIONS The homozygous GHRHR mutation was rare, being detected in only one Japanese IGHD family. Future research is needed to clarify the genetic contributions of heterozygous functional promoter variants to GHD, ISS and normal-stature variations.
Collapse
Affiliation(s)
- Hiroshi Inoue
- Division of Genetic Information, Institute for Genome Research, The University of Tokushima, Kuramoto 3-18-15, Tokushima 770-8503, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Iovino M, Triggiani V, Giagulli VA, Iovine N, Licchelli B, Resta F, Sabbà C, Tafaro E, Solimando A, Tommasicchio A, Guastamacchia E. Difference in growth hormone response to growth hormone-releasing hormone (GHRH) testing following GHRH subacute treatment in normal aging and growth hormone-deficient adults: possible perspectives for therapeutic use of GHRH or its analogs in elderly subjects? Immunopharmacol Immunotoxicol 2010; 33:334-7. [PMID: 20843274 DOI: 10.3109/08923973.2010.510844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The somatotroph axis function shows a decline in the elderly (somatopause). In particular growth hormone (GH) response to GH-releasing hormone (GHRH) is reduced in aged man but less than that observed in GH-deficient adults (GHDAs). Plasma GH response to GHRH (1 µg/kg BW) was significantly lower in four GHDAs than in seven healthy aged men 30, 60, and 90 min after acute GHRH administration. To verify whether a priming regimen might be able to increase the reduced GH response to GHRH, both healthy aged men and GHDA patients underwent repetitive administration of GHRH (100 µg GHRH intravenously as a single morning dose, every 2 days for 12 days). After the GHRH-priming regimen, plasma GH values 30, 60, and 90 min after the acute GHRH test were significantly higher than values at the corresponding time points before priming regimen in healthy aged men but not in GHDA patients. These findings confirmed that somatotroph cells become less sensitive to GHRH with normal aging and demonstrate that repetitive administration of GHRH restores the attenuated response only in healthy aged men but not in GHDA patients. This could support the possible use of GHRH or its analogs instead of recombinant human GH in elderly patients with the advantage of preserving the endogenous pulses of GH with the secretion of the different isoforms of GH. However, concerns arise about the possible role of these molecules in tumorigenesis and tumor growth promotion.
Collapse
Affiliation(s)
- M Iovino
- Endocrinology, General Hospital of Eboli, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Unal EB, Gursoy A, Erman B. VitAL: Viterbi algorithm for de novo peptide design. PLoS One 2010; 5:e10926. [PMID: 20532195 PMCID: PMC2880006 DOI: 10.1371/journal.pone.0010926] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 05/07/2010] [Indexed: 01/18/2023] Open
Abstract
Background Drug design against proteins to cure various diseases has been studied for several years. Numerous design techniques were discovered for small organic molecules for specific protein targets. The specificity, toxicity and selectivity of small molecules are hard problems to solve. The use of peptide drugs enables a partial solution to the toxicity problem. There has been a wide interest in peptide design, but the design techniques of a specific and selective peptide inhibitor against a protein target have not yet been established. Methodology/Principal Findings A novel de novo peptide design approach is developed to block activities of disease related protein targets. No prior training, based on known peptides, is necessary. The method sequentially generates the peptide by docking its residues pair by pair along a chosen path on a protein. The binding site on the protein is determined via the coarse grained Gaussian Network Model. A binding path is determined. The best fitting peptide is constructed by generating all possible peptide pairs at each point along the path and determining the binding energies between these pairs and the specific location on the protein using AutoDock. The Markov based partition function for all possible choices of the peptides along the path is generated by a matrix multiplication scheme. The best fitting peptide for the given surface is obtained by a Hidden Markov model using Viterbi decoding. The suitability of the conformations of the peptides that result upon binding on the surface are included in the algorithm by considering the intrinsic Ramachandran potentials. Conclusions/Significance The model is tested on known protein-peptide inhibitor complexes. The present algorithm predicts peptides that have better binding energies than those of the existing ones. Finally, a heptapeptide is designed for a protein that has excellent binding affinity according to AutoDock results.
Collapse
Affiliation(s)
- E. Besray Unal
- Center for Computational Biology and Bioinformatics, Koc University, Istanbul, Turkey
| | - Attila Gursoy
- Center for Computational Biology and Bioinformatics, Koc University, Istanbul, Turkey
| | - Burak Erman
- Center for Computational Biology and Bioinformatics, Koc University, Istanbul, Turkey
- * E-mail:
| |
Collapse
|
41
|
Abstract
Many theories aim at explaining the mechanisms of aging and death in humans. Decreased levels of androgens, growth hormone, and insulin-like growth factor accompany natural aging in men. Androgens influence the growth and maturation of men in various stages of their life. The action of androgens is performed by binding or not binding to androgen receptors. However, various actions of androgens were clarified after the discovery and genotyping of the androgen receptor. The influence of androgens on the lipid profile was reported by several researchers. This negative influence of androgens in men and the positive influence of estrogens in women are responsible for the higher impact of atherogenesis in men compared with women. In aging men, this negative influence of androgens on the lipid profile is more pronounced. This review considers the influence of age on lipid metabolism in men.
Collapse
|
42
|
Effects of genetic variability of the dairy goat growth hormone releasing hormone receptor (GHRHR) gene on growth traits. Mol Biol Rep 2010; 38:539-44. [PMID: 20354904 DOI: 10.1007/s11033-010-0138-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Accepted: 03/23/2010] [Indexed: 10/19/2022]
Abstract
Growth hormone-releasing hormone receptor (GHRHR) plays a critical role in growth hormone (GH) synthesis, release and regulation of pituitary somatotroph expansion in vertebrates. The objective of this study was to investigate variations in goat GHRHR gene and their associations with growth traits in 668 dairy goats. The results showed four novel single nucleotide polymorphisms (SNPs): NC_007302:g.5203C>T, 7307C>G, 9583G>A and 9668A>C. In detail, the novel SNP C>T in the 5203rd nucleotide identified a missense mutation: CCC (Pro)>TCC (Phe) at position 116aa of the goat GHRHR (423aa). Besides, 9583G>A and 9668A>C polymorphism were in complete linkage disequilibrium. The genetic diversity analysis revealed that the Guanzhong dairy goat possessed intermediate genetic diversity in P3 and P7 loci, and the Xinong Sannen dairy goat belonged to poor genetic diversity in P4 locus. Significant associations between the genotypes of P3 locus and body length, body height and chest circumference was observed in Guanzhong goat (P<0.05). However, in Xinong saanen population, significant statistical difference was only found in body height and body length (P<0.05). In P4 and P7 loci, no significant associations were detected between any variant sites and body length, body height and chest circumference, as well as for the milk traits (P>0.05). These results strongly suggested that the goat GHRHR gene is a candidate gene that influences growth traits in dairy goat.
Collapse
|
43
|
Granata R, Trovato L, Gallo MP, Destefanis S, Settanni F, Scarlatti F, Brero A, Ramella R, Volante M, Isgaard J, Levi R, Papotti M, Alloatti G, Ghigo E. Growth hormone-releasing hormone promotes survival of cardiac myocytes in vitro and protects against ischaemia-reperfusion injury in rat heart. Cardiovasc Res 2009; 83:303-12. [PMID: 19293247 DOI: 10.1093/cvr/cvp090] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIMS The hypothalamic neuropeptide growth hormone-releasing hormone (GHRH) stimulates GH synthesis and release in the pituitary. GHRH also exerts proliferative effects in extrapituitary cells, whereas GHRH antagonists have been shown to suppress cancer cell proliferation. We investigated GHRH effects on cardiac myocyte cell survival and the underlying signalling mechanisms. METHODS AND RESULTS Reverse transcriptase-polymerase chain reaction analysis showed GHRH receptor (GHRH-R) mRNA in adult rat ventricular myocytes (ARVMs) and in rat heart H9c2 cells. In ARVMs, GHRH prevented cell death and caspase-3 activation induced by serum starvation and by the beta-adrenergic receptor agonist isoproterenol. The GHRH-R antagonist JV-1-36 abolished GHRH survival action under both experimental conditions. GHRH-induced cardiac cell protection required extracellular signal-regulated kinase (ERK)1/2 and phosphoinositide-3 kinase (PI3K)/Akt activation and adenylyl cyclase/cAMP/protein kinase A signalling. Isoproterenol strongly upregulated the mRNA and protein of the pro-apoptotic inducible cAMP early repressor, whereas GHRH completely blocked this effect. Similar to ARVMs, in H9c2 cardiac cells, GHRH inhibited serum starvation- and isoproterenol-induced cell death and apoptosis through the same signalling pathways. Finally, GHRH improved left ventricular recovery during reperfusion and reduced infarct size in Langendorff-perfused rat hearts, subjected to ischaemia-reperfusion (I/R) injury. These effects involved PI3K/Akt signalling and were inhibited by JV-1-36. CONCLUSION Our findings suggest that GHRH promotes cardiac myocyte survival through multiple signalling mechanisms and protects against I/R injury in isolated rat heart, indicating a novel cardioprotective role of this hormone.
Collapse
Affiliation(s)
- Riccarda Granata
- Laboratory of Molecular and Cellular Endocrinology, Department of Internal Medicine, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wang Y, Tomlinson B. Tesamorelin, a human growth hormone releasing factor analogue. Expert Opin Investig Drugs 2009; 18:303-10. [DOI: 10.1517/13543780802707658] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
45
|
Chapter 3 Diseases Associated with Growth Hormone‐Releasing Hormone Receptor (GHRHR) Mutations. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 88:57-84. [DOI: 10.1016/s1877-1173(09)88003-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
46
|
Luque RM, Huang ZH, Shah B, Mazzone T, Kineman RD. Effects of leptin replacement on hypothalamic-pituitary growth hormone axis function and circulating ghrelin levels in ob/ob mice. Am J Physiol Endocrinol Metab 2007; 292:E891-9. [PMID: 17122091 DOI: 10.1152/ajpendo.00258.2006] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Leptin-deficient obese mice (ob/ob) have decreased circulating growth hormone (GH) and pituitary GH and ghrelin receptor (GHS-R) mRNA levels, whereas hypothalamic GH-releasing hormone (GHRH) and somatostatin (SST) expression do not differ from lean controls. Given the fact that GH is suppressed in diet-induced obesity (a state of hyperleptinemia), it remains to be determined whether the absence of leptin contributes to changes in the GH axis of ob/ob mice. Therefore, to study the impact of leptin replacement on the hypothalamic-pituitary GH axis of ob/ob mice, leptin was infused for 7 days (sc), resulting in circulating leptin levels that were similar to wild-type controls (approximately 1 ng/ml). Leptin treatment reduced food intake, body weight, and circulating insulin while elevating circulating n-octanoyl ghrelin concentrations. Leptin treatment did not alter hypothalamic GHRH, SST, or GHS-R mRNA levels compared with vehicle-treated controls. However, leptin significantly increased pituitary GH and GHRH-R expression and tended to enhance circulating GH levels, but this latter effect did not reach statistical significance. In vitro, leptin (1 ng/ml, 24 h) did not affect pituitary GH, GHRH-R, or GHS-R mRNA but did enhance GH release. The in vivo effects of leptin on circulating hormone and pituitary mRNA levels were not replicated by pair feeding ob/ob mice to match the food intake of leptin-treated mice. However, leptin did prevent the fall in hypothalamic GHRH mRNA and circulating IGF-I levels observed in pair-fed mice. These results demonstrate that leptin replacement has positive effects on multiple levels of GH axis function in ob/ob mice.
Collapse
Affiliation(s)
- Raul M Luque
- Department of Medicine, University of Illinois at Chicago, and Jesse Brown VA Medical Center, Research and Development Division, M. P. 151, West Side, Suite no. 6215, 820 South Damen Ave., Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
47
|
Lutz L, Schoefield N, Crowe C, Dufourny L, Skinner DC. No effect of nutrient restriction from gestational days 28 to 78 on immunocytochemically detectable growth hormone-releasing hormone (GHRH) neurons and GHRH receptor colocalization in somatotropes of the ovine female fetus. J Chem Neuroanat 2007; 33:34-41. [PMID: 17134871 DOI: 10.1016/j.jchemneu.2006.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Revised: 10/19/2006] [Accepted: 10/26/2006] [Indexed: 11/28/2022]
Abstract
The maternal environment affects fetal development and may permanently affect the physiology of the adult. Fetal growth hormone (GH) secretion is increased by maternal undernutrition but the physiological mechanisms responsible for this increase are unknown. We have recently found evidence suggesting that the GHRH component of the fetal neuroendocrine GH axis may be perturbed by undernutrition. This study sought to determine the effect of maternal undernutrition on immunocytochemically detectable GHRH neurons and the expression of GHRH receptors by somatotropes in the pituitary gland. Ewes were grouped (n=12 per group) randomly into control (fed 100% of requirements) or nutrient restricted (fed 50% of requirements) from days 28 to 78 of gestation, corresponding to the period from implantation to the end of placentation. At day 78, half the ewes were killed and the fetal brains were perfused. The remaining ewes were re-alimented to 100% of nutritional requirements and killed at day 135. There was no effect of nutrition restriction or age on the number of GHRH neurons. Similarly, the mean density and percentage of somatotropes expressing GHRH receptors was not significantly different between treatment groups at either age. This study found no effect, as determined by immunocytochemistry, of nutrient restriction on the GHRH component of the fetal neuroendocrine GH axis. It remains to be established if the release of GHRH and responsiveness of somatotropes to GHRH in the fetus are affected by undernutrition.
Collapse
Affiliation(s)
- Lacey Lutz
- Department of Zoology and Physiology and Neurobiology Program, University of Wyoming, Laramie, WY 82071, USA
| | | | | | | | | |
Collapse
|
48
|
Wagner K, Hemminki K, Försti A. The GH1/IGF-1 axis polymorphisms and their impact on breast cancer development. Breast Cancer Res Treat 2006; 104:233-48. [PMID: 17082888 DOI: 10.1007/s10549-006-9411-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2006] [Accepted: 09/17/2006] [Indexed: 10/24/2022]
Abstract
The growth hormone 1/insulin-like growth factor-1 (GH1/IGF-1) axis plays an essential role in the development of the breast by regulating cell proliferation, differentiation and apoptosis. Imbalances within this axis lead to an aberrant signalling and recent research has focussed on the overexpression of these growth factors and their involvement in breast cancer development. The increased understanding of the molecular mechanisms and signalling pathways connected to the GH1/IGF-1 axis has provided important insights into aetiology, prevention and therapy for breast cancer. However, to identify the contribution of the GH1/IGF-1 signalling pathway to cancer risk still remains a challenge since the results of various studies are controversial. Here, we discuss the influence of low-penetrance polymorphisms in the genes along the GH1/IGF-1 axis and their impact on hormone levels and cancer risk, especially breast cancer. We point out what is known about the effects of the variants and show how the interaction of genetic variants affects breast cancer risk.
Collapse
Affiliation(s)
- Kerstin Wagner
- Division of Molecular Genetic Epidemiology C050, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany.
| | | | | |
Collapse
|
49
|
Cheong HS, Yoon DH, Kim LH, Park BL, Choi YH, Chung ER, Cho YM, Park EW, Cheong IC, Oh SJ, Yi SG, Park T, Shin HD. Growth hormone-releasing hormone (GHRH) polymorphisms associated with carcass traits of meat in Korean cattle. BMC Genet 2006; 7:35. [PMID: 16749938 PMCID: PMC1524984 DOI: 10.1186/1471-2156-7-35] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Accepted: 06/03/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cold carcass weight (CW) and longissimus muscle area (EMA) are the major quantitative traits in beef cattle. In this study, we found several polymorphisms of growth hormone-releasing hormone (GHRH) gene and examined the association of polymorphisms with carcass traits (CW and EMA) in Korean native cattle (Hanwoo). RESULTS By direct DNA sequencing in 24 unrelated Korean cattle, we identified 12 single nucleotide polymorphisms within the 9 kb full gene region, including the 1.5 kb promoter region. Among them, six polymorphic sites were selected for genotyping in our beef cattle (n = 428) and five marker haplotypes (frequency > 0.1) were identified. Statistical analysis revealed that -4241A>T showed significant associations with CW and EMA. CONCLUSION Our findings suggest that polymorphisms in GHRH might be one of the important genetic factors that influence carcass yield in beef cattle. Sequence variation/haplotype information identified in this study would provide valuable information for the production of a commercial line of beef cattle.
Collapse
Affiliation(s)
- Hyun Sub Cheong
- Department of Genetic Epidemiology, SNP Genetics, Inc., Seoul, 153-803, Korea
| | - Du-Hak Yoon
- National Livestock Research Institute, RDA, 441-706, Korea
| | - Lyoung Hyo Kim
- Department of Genetic Epidemiology, SNP Genetics, Inc., Seoul, 153-803, Korea
| | - Byung Lae Park
- Department of Genetic Epidemiology, SNP Genetics, Inc., Seoul, 153-803, Korea
| | - Yoo Hyun Choi
- Department of Genetic Epidemiology, SNP Genetics, Inc., Seoul, 153-803, Korea
| | - Eui Ryong Chung
- Department of Biotechnology, Sangi University, Wonju, Kangwon Do, 220-702, Korea
| | - Yong Min Cho
- National Livestock Research Institute, RDA, 441-706, Korea
| | - Eng Woo Park
- National Livestock Research Institute, RDA, 441-706, Korea
| | | | - Sung-Jong Oh
- National Livestock Research Institute, RDA, 441-706, Korea
| | - Sung-Gon Yi
- Department of Statistics, Seoul National University, Seoul, 151-747, Korea
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, 151-747, Korea
| | - Hyoung Doo Shin
- Department of Genetic Epidemiology, SNP Genetics, Inc., Seoul, 153-803, Korea
| |
Collapse
|
50
|
Lutz L, Dufourny L, Skinner DC. Effect of nutrient restriction on the somatotropes and substance P-immunoreactive cells in the pituitary of the female ovine fetus. Growth Horm IGF Res 2006; 16:108-118. [PMID: 16600649 DOI: 10.1016/j.ghir.2006.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 02/15/2006] [Accepted: 02/28/2006] [Indexed: 10/24/2022]
Abstract
The maternal environment affects fetal development and may influence the physiology of the adult. Fetal growth hormone (GH) is increased by maternal undernutrition but the mechanisms responsible are unknown. This study determined the effect of maternal undernutrition on the development of fetal pituitary somatotropes in the female. Ewes were grouped randomly into control (fed 100% of requirements) or nutrient restricted (fed 50%) from Days 28 to 78 of gestation. At Day 78, the ewes were killed and fetuses collected (Day 78 NR (nutrient restricted): n=6; Day 78C (control): n=6). Remaining ewes were realimented to 100% of nutritional requirements and were killed at Day 135 (Day 135 NR (nutrient restricted): n=6; Day 135 C (control): n=6). Somatotropes were visualized immunocytochemically and the size, mean density, total percentage and proportion colocalized with substance P were determined for each group. Nutrient restriction increased (p<0.01) the density of pituitary cells in Day 78 fetuses but this difference was no longer apparent by Day 135 after realimentation. The density and proportion of somatotropes were not different between treatment groups at Day 78 but were significantly (p<0.05) lower in the nutrient restricted Day 135 fetuses as compared to the Day 135 control animals. Somatotropes from restricted fetuses were significantly (p<0.001) larger at Day 78. Nutrient restriction increased the density (p<0.001) and percentage (p<0.05) of substance P-immunoreactive cells Day 135 fetuses. Similarly, the proportion of somatotropes that expressed substance P was significantly (p<0.05) increased by nutrient restriction in the Day 135 fetuses. Although nearly two thirds of substance P-immunoreactive cells co-expressed GH, there was no significant effect of treatment on this co-expression. Additional studies are required to determine if other components of the neuroendocrine GH axis are affected by this nutritional insult, if the alterations that we have observed, particularly in the tachykinin system, persist into adulthood and, importantly, what are the long-term consequences of an altered GH axis.
Collapse
Affiliation(s)
- Lacey Lutz
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY 82071, USA
| | | | | |
Collapse
|