1
|
Montero-Hidalgo AJ, Del Rio-Moreno M, Pérez-Gómez JM, Luque RM, Kineman RD. Update on regulation of GHRH and its actions on GH secretion in health and disease. Rev Endocr Metab Disord 2025:10.1007/s11154-025-09943-y. [PMID: 39838154 DOI: 10.1007/s11154-025-09943-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2025] [Indexed: 01/23/2025]
Abstract
This review focuses on our current understanding of how growth hormone releasing hormone (GHRH): 1) stimulates GH release and synthesis from pituitary growth hormone (GH)-producing cells (somatotropes), 2) drives somatotrope proliferation, 3) is negatively regulated by somatostatin (SST), GH and IGF1, 4) is altered throughout lifespan and in response to metabolic challenges, and 5) analogues can be used clinically to treat conditions of GH excess or deficiency. Although a large body of early work provides an underpinning for our current understanding of GHRH, this review specifically highlights more recent work that was made possible by state-of-the-art analytical tools, receptor-specific agonists and antagonists, high-resolution in vivo and ex vivo imaging and the development of tissue (cell) -specific ablation mouse models, to paint a more detailed picture of the regulation and actions of GHRH.
Collapse
Affiliation(s)
- Antonio J Montero-Hidalgo
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
| | - Mercedes Del Rio-Moreno
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown Veterans Affairs Medical Center, Research and Development Division Chicago, 820 S. Damen Ave., MP151, Rm 6215, Chicago, IL, USA
| | - Jesús M Pérez-Gómez
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de La Obesidad y Nutrición, Cordoba, CIBERobn, Spain
| | - Rhonda D Kineman
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA.
- Jesse Brown Veterans Affairs Medical Center, Research and Development Division Chicago, 820 S. Damen Ave., MP151, Rm 6215, Chicago, IL, USA.
| |
Collapse
|
2
|
Boan AF, Delgadin TH, Canosa LF, Fernandino JI. Loss of function in somatostatin receptor 5 has no impact on the growth of medaka fish due to compensation by the other paralogs. Gen Comp Endocrinol 2024; 351:114478. [PMID: 38412943 DOI: 10.1016/j.ygcen.2024.114478] [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: 09/15/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 02/29/2024]
Abstract
Somatic growth in vertebrates is regulated endocrinologically by the somatotropic axis, headed by the growth hormone (GH) and the insulin growth factor-I (IGF-I). Somatostatin (Sst), a peptide hormone synthesized in the hypothalamus, modulates GH actions through its receptors (Sstr). Four Sstr subtypes (Sstr 1-3 and 5) have been identified in teleosts. However, little is known about whether they have a specific function or tissue expression. The aim of this study was to determine the role of sstr2 and sstr5 in the growth of the medaka (Oryzias latipes). The assessed expression pattern across diverse tissues highlighted greater prevalence of sstr1 and sstr3 in brain, intestine and muscle than in pituitary or liver. The expression of sstr2 was high in all the tissues tested, while sstr5 was predominantly expressed in the pituitary gland. A CRISPR/Cas9 sstr5 mutant with loss of function (sstr5-/-) was produced. Assessment of sstr5-/- indicated no significant difference with the wild type regarding growth parameters such as standard length, body depth, or peduncle depth. Furthermore, the functional loss of sstr5 had no impact on the response to a nutritional challenge. The fact that several sstr subtypes were upregulated in different tissues in sstr5-/- medaka suggests that in the mutant fish, there may be a compensatory effect on the different tissues, predominantly by sstr1 in the liver, brain and pituitary, with sstr2 being upregulated in pituitary and liver, and sstr3 only presenting differential expression in the brain. Analysis of the sstr subtype and the sstr5-/- fish showed that sstr5 was not the only somatostatin receptor responsible for Sst-mediated Gh regulation.
Collapse
Affiliation(s)
- A F Boan
- Instituto Tecnológico de Chascomús, INTECH (CONICET-UNSAM), Chascomús, Argentina; Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Argentina
| | - T H Delgadin
- Universidad Nacional del Santa, Nuevo Chimbote, Ancash, Perú
| | - L F Canosa
- Instituto Tecnológico de Chascomús, INTECH (CONICET-UNSAM), Chascomús, Argentina; Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Argentina
| | - J I Fernandino
- Instituto Tecnológico de Chascomús, INTECH (CONICET-UNSAM), Chascomús, Argentina; Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Argentina.
| |
Collapse
|
3
|
Sada V, Puliani G, Feola T, Pirchio R, Pofi R, Sesti F, De Alcubierre D, Amodeo ME, D'Aniello F, Vincenzi L, Gianfrilli D, Isidori AM, Grossman AB, Sbardella E. Tall stature and gigantism in transition age: clinical and genetic aspects-a literature review and recommendations. J Endocrinol Invest 2024; 47:777-793. [PMID: 37891382 DOI: 10.1007/s40618-023-02223-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023]
Abstract
PURPOSE Tall stature is defined as height greater than the threshold of more than 2 standard deviations above the average population height for age, sex, and ethnicity. Many studies have described the main aspects of this condition during puberty, but an analysis of the characteristics that the physician should consider in the differential diagnosis of gigantism-tall stature secondary to a pituitary tumour-during the transition age (15-25 years) is still lacking. METHODS A comprehensive search of English-language original articles was conducted in the MEDLINE database (December 2021-March 2022). We selected all studies regarding epidemiology, genetic aspects, and the diagnosis of tall stature and gigantism during the transition age. RESULTS Generally, referrals for tall stature are not as frequent as expected because most cases are familial and are usually unreported by parents and patients to endocrinologists. For this reason, lacking such experience of tall stature, familiarity with many rarer overgrowth syndromes is essential. In the transition age, it is important but challenging to distinguish adolescents with high constitutional stature from those with gigantism. Pituitary gigantism is a rare disease in the transition age, but its systemic complications are very relevant for future health. Endocrine evaluation is crucial for identifying conditions that require hormonal treatment so that they can be treated early to improve the quality of life and prevent comorbidities of individual patient in this age range. CONCLUSION The aim of our review is to provide a practical clinical approach to recognise adolescents, potentially affected by gigantism, as early as possible.
Collapse
Affiliation(s)
- V Sada
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - G Puliani
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - T Feola
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli, Italy
| | - R Pirchio
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Degli Studi di Napoli "Federico II", Naples, Italy
| | - R Pofi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford University Hospitals, NHS Trust, Oxford, UK
| | - F Sesti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - D De Alcubierre
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - M E Amodeo
- Dipartimento Pediatrico Universitario Ospedaliero, Bambino Gesù Children Hospital, Rome, Italy
| | - F D'Aniello
- Dipartimento Pediatrico Universitario Ospedaliero, Bambino Gesù Children Hospital, Rome, Italy
| | - L Vincenzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - D Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - A M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Centre for Rare Diseases (ENDO-ERN Accredited), Policlinico Umberto I, Rome, Italy
| | - A B Grossman
- Green Templeton College, University of Oxford, Oxford, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
| | - E Sbardella
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
4
|
Cao L, Zhou J, Ma W, Zhang H, Pan H, Xu M, Wang Y, Wang P, Xiang X, Liu Y, Qiu X, Zhou X, Wang X. Identification of lncRNA-based regulatory mechanisms of Takifugu rubripes growth traits in fast and slow-growing family lines. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 49:101164. [PMID: 37976965 DOI: 10.1016/j.cbd.2023.101164] [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: 08/16/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
Family selection is an important method in fish aquaculture because growth is the most important economic trait. Fast-and slow-growing families of tiger puffer fish (Takifugu rubripes) have been established through family selection. The development of teleost fish is primarily controlled by the growth hormone (GH)-insulin-like growth factor 1 (IGF-1) axis that includes the hypothalamus-pituitary-liver. In this study, the molecular mechanisms underlying T. rubripes growth were analyzed by comparing transcriptomes from fast- and slow-growing families. The expressions of 214 lncRNAs were upregulated, and those of 226 were downregulated in the brain tissues of the fast-growing T. rubripes family compared to those of the slow-growing family. Differentially expressed lncRNAs centrally regulate mitogen-activated protein kinase (MAPK) and forkhead box O (FoxO) signaling pathways. Based on the results of lncRNA-gene network construction, we found that lncRNA3133.13, lncRNA23169.1, lncRNA23145.1, and lncRNA23141.3 regulated all four genes (igf1, mdm2, flt3, and cwf19l1). In addition, lncRNA7184.10 may be a negative regulator of rasgrp2 and a positive regulator of gadd45ga, foxo3b, and dusp5. These target genes are associated with the growth and development of organisms through the PI3K/AKT and MAPK/ERK pathways. Overall, transcriptomic analyses of fast- and slow-growing families of T. rubripes provided insights into the molecular mechanisms of teleost fish growth rates. Further, these analyses provide evidence for key genes related to growth regulation and the lncRNA expression regulatory network that will provide a framework for improving puffer fish germplasm resources.
Collapse
Affiliation(s)
- Lirong Cao
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Jinxu Zhou
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Wenchao Ma
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Huakun Zhang
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Hanbai Pan
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Mingjie Xu
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Yusen Wang
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Peiyang Wang
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Xuejian Xiang
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Yang Liu
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Xuemei Qiu
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Xiaoxu Zhou
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China.
| | - Xiuli Wang
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Pufferfish Breeding and Culture in Liaoning Province, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
5
|
Przybył B, Wójcik-Gładysz A, Gajewska A, Szlis M. Brain-derived neurotrophic factor (BDNF) affects somatotrophic
axis activity in sheep. JOURNAL OF ANIMAL AND FEED SCIENCES 2021. [DOI: 10.22358/jafs/143353/2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
6
|
Giglione E, Lapolla R, Cianfarani S, Faienza MF, Fintini D, Weber G, Delvecchio M, Valerio G. Linear growth and puberty in childhood obesity: what is new? Minerva Pediatr (Torino) 2021; 73:563-571. [PMID: 34309346 DOI: 10.23736/s2724-5276.21.06543-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pediatric obesity is a growing and alarming global health problem and represents an important determinant of morbidity. Since nutrition plays an important role in regulating growth and development, the excess weight gain related to overnutrition can affect growth patterns, bone maturation and pubertal development. The purpose of this review is to summarize the current knowledge about the effect of primary obesity on linear growth and pubertal development in children and adolescents. Evidences about regulatory hormones and adipokines that may be involved in the physiology of childhood growth in the context of obesity were also discussed. The most recent literature confirms previous studies indicating that linear growth is accelerated (mainly due to longer trunks rather than longer legs) and bone age is advanced in prepubertal children with obesity, while there is a reduction of pubertal height gain and attainment of normal adult height. Conflicting results are reported on the timing of puberty, specifically in boys. Indeed, previous studies suggested earlier onset of puberty in obese girls and overweight boys, and a delayed puberty in obese boys. Conversely, the most recent studies show more consistently an earlier onset and completion of pubertal development also in boys with obesity. Considering the false belief of health associated with transient taller stature in children and the adverse outcomes related to early puberty, interventions on diet and physical activity are urgently needed to tackle the epidemics of childhood obesity in public health and clinical setting.
Collapse
Affiliation(s)
| | - Rosa Lapolla
- Dipartimento Materno-Infantile, AOR San Carlo, Potenza, Italy
| | - Stefano Cianfarani
- Dipartimento Pediatrico Universitario Ospedaliero, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Department of Women's and Children's Health, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Maria F Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University A. Moro, Bari, Italy
| | - Danilo Fintini
- Endocrinology Unit, Pediatric University Department, Bambino Gesù Children Hospital, Rome, Italy
| | - Giovanna Weber
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
| | - Maurizio Delvecchio
- Metabolic Disorders and Genetic Unit, Giovanni XXIII Children Hospital, Bari, Italy
| | - Giuliana Valerio
- Department of Movement Sciences and Wellbeing, University of Naples Parthenope, Naples, Italy -
| |
Collapse
|
7
|
Montjean R, Escaich S, Paolini R, Carelli C, Pirson S, Neutelings T, Henrotin Y, Vêtu C. REG-O3 chimeric peptide combining growth hormone and somatostatin sequences improves joint function and prevents cartilage degradation in rat model of traumatic knee osteoarthritis. PLoS One 2020; 15:e0231240. [PMID: 32287299 PMCID: PMC7156079 DOI: 10.1371/journal.pone.0231240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
Objective REG-O3 is a 24-aminoacid chimeric peptide combining a sequence derived from growth hormone (GH) and an analog of somatostatin (SST), molecules displaying cartilage repair and anti-inflammatory properties, respectively. This study aimed to investigate the disease-modifying osteoarthritis drug (DMOAD) potential of REG-O3 by analyzing its effect on pain, joint function and structure, upon injection into osteoarthritic rat knee joint. Design Osteoarthritis was induced in the right knee of mature male Lewis rats (n = 12/group) by surgical transection of the anterior cruciate ligament (ACLT) combined with partial medial meniscectomy (pMMx). Treatments were administered intra-articularly from fourteen days after surgery through three consecutive injections one week apart. The effect of REG-O3, solubilized in a liposomal solution and injected at either 5, 25 or 50 μg/50 μL, was compared to liposomal (LIP), dexamethasone and hyaluronic acid (HA) solutions. The study endpoints were the pain/function measured once a week throughout the entire study, and the joint structure evaluated eight weeks after surgery using OARSI score. Results ACLT/pMMx surgery induced a significant modification of weight bearing in all groups. When compared to liposomal solution, REG-O3 was able to significantly improve weight bearing as efficiently as dexamethasone and HA. REG-O3 (25 μg) was also able to significantly decrease OARSI histological global score as well as degeneration of both cartilage and matrix while the other treatments did not. Conclusion This study provides evidence of a remarkable protecting effect of REG-O3 on pain/knee joint function and cartilage/matrix degradation in ACLT/pMMx model of rat osteoarthritis. REG-O3 thus displays an interesting profile as a DMOAD.
Collapse
Affiliation(s)
| | - Sonia Escaich
- Regulaxis SAS, Romainville, France
- ESE Conseil, Saint-Cloud, France
| | | | | | | | | | - Yves Henrotin
- Artialis SA, Tour GIGA, CHU Sart-Tilman, Liège, Belgium
- Bone and Cartilage Research Unit, Arthropôle Liège, University of Liège, Liège, Belgium
- Physical Therapy and Rehabilitation Department, Princess Paola Hospital, Vivalia, Marche-en-Famenne, Belgium
- * E-mail:
| | | |
Collapse
|
8
|
Van Dyck L, Derese I, Vander Perre S, Wouters PJ, Casaer MP, Hermans G, Van den Berghe G, Vanhorebeek I. The GH Axis in Relation to Accepting an Early Macronutrient Deficit and Outcome of Critically Ill Patients. J Clin Endocrinol Metab 2019; 104:5507-5518. [PMID: 31361307 DOI: 10.1210/jc.2019-00842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/24/2019] [Indexed: 02/13/2023]
Abstract
CONTEXT Changes in the GH axis during critical illness resemble fasting in healthy adults and contribute to hypercatabolism, which potentially affects outcome. Accepting macronutrient deficits by withholding parenteral nutrition (PN) during the first week in the intensive care unit (ICU; late PN) reduced complications and accelerated recovery as compared with early use of PN (early PN). OBJECTIVE To investigate how late PN affects the GH axis in relation to its clinical outcome benefits. DESIGN Preplanned subanalysis of the Early Parenteral Nutrition Completing Enteral Nutrition in Adult Critically Ill Patients randomized controlled trial. PARTICIPANTS A total of 1128 patients for time-course study, 20 patients investigated for nocturnal GH pulsatility, and 600 patients investigated for muscle weakness, with early PN and late PN patients having comparable baseline characteristics. INTERVENTION Withholding PN during the first ICU week (late PN) vs early PN. MAIN OUTCOME MEASURES Changes in serum GH, IGF-I, IGF-binding protein (IGFBP) 3, and IGFBP1 concentrations from ICU admission to day 4 or last ICU day for patients with a shorter ICU stay (d4/LD) and association in multivariable analyses with likelihood of earlier live ICU discharge, risk of new infection, and muscle weakness. RESULTS Late PN attenuated a rise in serum GH and IGF-I (P < 0.0001), did not affect IGFBP3, and attenuated a decrease in IGFBP1 concentrations from admission to d4/LD (P < 0.0001) as compared with early PN. Late PN decreased nonpulsatile (P = 0.005), but not pulsatile, GH secretion. Adjusting the multivariable models for the observed GH axis alterations increased the independent benefit of late PN for all outcomes. GH axis alterations induced by late PN were independently associated with adverse outcomes (P ≤ 0.03). CONCLUSION Accepting macronutrient deficits early during critical illness further suppressed the GH axis, which statistically attenuated its clinical outcome benefits.
Collapse
Affiliation(s)
- Lisa Van Dyck
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Inge Derese
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Sarah Vander Perre
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Pieter J Wouters
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Michaël P Casaer
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Greet Hermans
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Greet Van den Berghe
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ilse Vanhorebeek
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Shalitin S, Kiess W. Putative Effects of Obesity on Linear Growth and Puberty
. Horm Res Paediatr 2018; 88:101-110. [PMID: 28183093 DOI: 10.1159/000455968] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/10/2017] [Indexed: 01/02/2023] Open
Abstract
Childhood obesity is a major public health problem that has grown to epidemic proportions throughout the world. Obesity is influenced by genetic and environmental factors. The nutritional status plays an important role in growth and body weight regulation. Excess adiposity during childhood can affect the process of growth and puberty. Obese children are frequently tall for their age, with accelerated epiphyseal growth plate maturation despite low growth hormone levels. Several regulatory hormones may affect the process of linear growth in the constellation of obesity, as high levels of insulin and leptin are observed in obese children. Leptin can act as a skeletal growth factor, with a direct effect on skeletal growth centers. The finding that overweight children, especially girls, tend to mature earlier than lean children has led to the hypothesis that the degree of body fatness may trigger the neuroendocrine events that lead to the onset of puberty. Leptin receptors have been identified in the hypothalamus, as well as in gonadotrope cells, ovarian follicular cells, and Leydig cells. The increased leptin and androgen levels seen in obese children may be implicated in their earlier onset of puberty and accelerated pubertal growth. This review is focused on the interaction between childhood obesity and growth and pubertal processes.
.
Collapse
Affiliation(s)
- Shlomit Shalitin
- The Jesse Z. and Sara Lea Shafer Institute of Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Wieland Kiess
- Department of Women and Child Health, Hospital for Children and Adolescents, University Hospitals, University of Leipzig, Leipzig, Germany
| |
Collapse
|
10
|
Dufourny L, Delmas O, Teixeira-Gomes AP, Decourt C, Sliwowska JH. Neuroanatomical connections between kisspeptin neurones and somatostatin neurones in female and male rat hypothalamus: a possible involvement of SSTR1 in kisspeptin release. J Neuroendocrinol 2018; 30:e12593. [PMID: 29543369 DOI: 10.1111/jne.12593] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/09/2018] [Indexed: 01/23/2023]
Abstract
Somatostatin (SST) a neuropeptide involved in the central modulation of several physiological functions, is co-distributed in the same hypothalamic areas as kisspeptin (KP), the most potent secretagogue of the gonadotropin-releasing hormone (GnRH) secretion known to date. As SST infused intracerebroventricularly (icv) evoked a potent inhibition of GnRH release, we explored neuroanatomical relationships between KP and SST populations in male and female rats. For that, intact males and ovariectomised oestradiol-replaced females were killed and their brains processed in order to simultaneously detect KP, SST and synapsin, a marker for synapses. We observed numerous appositions of KP on SST neurones both in female and male arcuate nucleus (ARC) and ventromedial hypothalamus. A large association between SST terminals and KP neurones at the level of the pre-optic area (POA) was also observed in female rats and in a more limited frame in males. Finally, most KP neurones from the ARC showed SST appositions in both sexes. To determine whether SST could affect KP cell activity, we assessed whether SST receptors (SSTR) were present on KP neurones in the ARC. We also looked for the presence of SSTR1 and SSTR2A in the brain of male rats. Brains were processed through a sequential double immunocytochemistry in order to detect KP and SSTR1 or KP and SSTR2A. We observed overlapping distributions of immunoreactive neurones for SSTR1 and KP and counted approximately one third of KP neurones with SSTR1. In contrast, neurones labelled for SSTR2A or KP were often juxtaposed in the ARC and the occurrence of double-labelled neurones was sporadic (<5%). These results suggest that SST action on KP neurones would pass mainly through SSTR1 at the level of the ARC. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laurence Dufourny
- UMR85 Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France
- CNRS, UMR 7247, F-37380, Nouzilly, France
- Université de Tours, F-37041, Tours, France
- IFCE, F-37380, Nouzilly, France
| | - Oona Delmas
- UMR85 Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France
- CNRS, UMR 7247, F-37380, Nouzilly, France
- Université de Tours, F-37041, Tours, France
- IFCE, F-37380, Nouzilly, France
| | - Ana-Paula Teixeira-Gomes
- UMR85 Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France
- CNRS, UMR 7247, F-37380, Nouzilly, France
- Université de Tours, F-37041, Tours, France
- IFCE, F-37380, Nouzilly, France
- INRA UMR INRA 1282 Infectiologie et Santé Publique, Université François Rabelais, F-37380, Nouzilly, France
| | - Caroline Decourt
- UMR85 Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France
- CNRS, UMR 7247, F-37380, Nouzilly, France
- Université de Tours, F-37041, Tours, France
- IFCE, F-37380, Nouzilly, France
| | - Joanna H Sliwowska
- Lab. of Neurobiology, Dpt of Veterinary Medicine and Animal Sciences, Poznan University of Life Science, 60-625, Poznan, Poland
| |
Collapse
|
11
|
Obestatin stimulates the somatotrophic axis activity in sheep. Brain Res 2018; 1678:278-287. [DOI: 10.1016/j.brainres.2017.10.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/25/2017] [Accepted: 10/31/2017] [Indexed: 01/12/2023]
|
12
|
Tuersunjiang N, Odhiambo JF, Shasa DR, Smith AM, Nathanielsz PW, Ford SP. Maternal obesity programs reduced leptin signaling in the pituitary and altered GH/IGF1 axis function leading to increased adiposity in adult sheep offspring. PLoS One 2017; 12:e0181795. [PMID: 28771488 PMCID: PMC5542597 DOI: 10.1371/journal.pone.0181795] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/09/2017] [Indexed: 11/29/2022] Open
Abstract
Studies in rodents highlight a role for leptin in stimulation of pituitary growth hormone (GH) secretion, with an impact on body composition regulation. We have reported that maternal obesity (MO) during ovine pregnancy results in hyperphagia, glucose-insulin dysregulation, increased adiposity, hypercortisolemia and hyperleptinemia in mature offspring subjected to a bout of ad libitum feeding. We hypothesized that MO reduces leptin signaling in the pituitary and down regulates the GH/IGF1 axis and increases circulating cortisol leading to increased adiposity in their adult offspring. Male lambs born to MO (n = 6) or control (CON, n = 6) ewes were fed only to requirements until placed on a 12 week ad libitum feeding trial at maturity. The pituitary, hypothalamic arcuate nucleus, and liver were collected at necropsy and mRNA and protein expression determined. Plasma cortisol concentrations were increased (P<0.05) in MO vs. CON offspring at the end of the feeding trial. Further, serum concentrations of IGF1 decreased (P<0.01) and GH tended to decrease (P<0.08) in MO vs. CON offspring. Pituitary mRNA and leptin receptor protein expression were decreased in MO vs. CON offspring in association with decreased GH mRNA expression, and decreased IGF1 mRNA and protein expression in liver. Liver 11β-hydroxysteroid dehydrogenase 1 (11βHSD1) expression was increased (P<0.01) and its cofactor hexose-6-phosphate dehydrogenase tended to increase (P<0.06) in MO vs. CON offspring. 11βHSD2 expression remained unchanged. These data indicate that MO induced an increase in liver conversion of cortisone to cortisol in adult offspring and support a role for leptin signaling in the pituitary in mediating offspring adiposity.
Collapse
Affiliation(s)
- Nuermaimaiti Tuersunjiang
- Center for the Study of Fetal Programming, Department of Animal Science, University of Wyoming, Laramie, Wyoming, United States of America
| | - John F. Odhiambo
- Center for the Study of Fetal Programming, Department of Animal Science, University of Wyoming, Laramie, Wyoming, United States of America
| | - Desiree R. Shasa
- Center for the Study of Fetal Programming, Department of Animal Science, University of Wyoming, Laramie, Wyoming, United States of America
| | - Ashley M. Smith
- Center for the Study of Fetal Programming, Department of Animal Science, University of Wyoming, Laramie, Wyoming, United States of America
| | - Peter W. Nathanielsz
- Center for the Study of Fetal Programming, Department of Animal Science, University of Wyoming, Laramie, Wyoming, United States of America
| | - Stephen P. Ford
- Center for the Study of Fetal Programming, Department of Animal Science, University of Wyoming, Laramie, Wyoming, United States of America
| |
Collapse
|
13
|
Obesity- and gender-dependent role of endogenous somatostatin and cortistatin in the regulation of endocrine and metabolic homeostasis in mice. Sci Rep 2016; 6:37992. [PMID: 27901064 PMCID: PMC5128804 DOI: 10.1038/srep37992] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/01/2016] [Indexed: 12/13/2022] Open
Abstract
Somatostatin (SST) and cortistatin (CORT) regulate numerous endocrine secretions and their absence [knockout (KO)-models] causes important endocrine-metabolic alterations, including pituitary dysregulations. We have demonstrated that the metabolic phenotype of single or combined SST/CORT KO-models is not drastically altered under normal conditions. However, the biological actions of SST/CORT are conditioned by the metabolic-status (e.g. obesity). Therefore, we used male/female SST- and CORT-KO mice fed low-fat (LF) or high-fat (HF) diet to explore the interplay between SST/CORT and obesity in the control of relevant pituitary-axes and whole-body metabolism. Our results showed that the SST/CORT role in the control of GH/prolactin secretions is maintained under LF- and HF-diet conditions as SST-KOs presented higher GH/prolactin-levels, while CORT-KOs displayed higher GH- and lower prolactin-levels than controls under both diets. Moreover, the impact of lack of SST/CORT on the metabolic-function was gender- and diet-dependent. Particularly, SST-KOs were more sensitive to HF-diet, exhibiting altered growth and body-composition (fat/lean percentage) and impaired glucose/insulin-metabolism, especially in males. Conversely, only males CORT-KO under LF-diet conditions exhibited significant alterations, displaying higher glucose-levels and insulin-resistance. Altogether, these data demonstrate a tight interplay between SST/CORT-axis and the metabolic status in the control of endocrine/metabolic functions and unveil a clear dissociation of SST/CORT roles.
Collapse
|
14
|
Han X, Li J, Cao X, Du X, Meng F, Zeng X. Surgical castration but not immuncastration is associated with reduced hypothalamic GnIH and GHRH/GH/IGF-I axis function in male rats. Theriogenology 2016; 86:657-65. [DOI: 10.1016/j.theriogenology.2016.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/13/2015] [Accepted: 02/22/2016] [Indexed: 10/22/2022]
|
15
|
Cordoba-Chacón J, Gahete MD, Pozo-Salas AI, de Lecea L, Castaño JP, Luque RM. Cortistatin Is a Key Factor Regulating the Sex-Dependent Response of the GH and Stress Axes to Fasting in Mice. Endocrinology 2016; 157:2810-23. [PMID: 27175972 DOI: 10.1210/en.2016-1195] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cortistatin (CORT) shares high structural and functional similarities with somatostatin (SST) but displays unique sex-dependent pituitary actions. Indeed, although female CORT-knockout (CORT-KO) mice exhibit enhanced GH expression/secretion, Proopiomelanocortin expression, and circulating ACTH/corticosterone/ghrelin levels, male CORT-KO mice only display increased plasma GH/corticosterone levels. Changes in peripheral ghrelin and SST (rather than hypothalamic levels) seem to regulate GH/ACTH axes in CORT-KOs under fed conditions. Because changes in GH/ACTH axes during fasting provide important adaptive mechanisms, we sought to determine whether CORT absence influences GH/ACTH axes during fasting. Accordingly, fed and fasted male/female CORT-KO were compared with littermate controls. Fasting increased circulating GH levels in male/female controls but not in CORT-KO, suggesting that CORT can be a relevant regulator of GH secretion during fasting. However, GH levels were already higher in CORT-KO than in controls in fed state, which might preclude a further elevation in GH levels. Interestingly, although fasting-induced pituitary GH expression was elevated in both male/female controls, GH expression only increased in fasted female CORT-KOs, likely owing to specific changes observed in key factors controlling somatotrope responsiveness (ie, circulating ghrelin and IGF-1, and pituitary GHRH and ghrelin receptor expression). Fasting increased corticosterone levels in control and, most prominently, in CORT-KO mice, which might be associated with a desensitization to SST signaling and to an augmentation in CRH and ghrelin-signaling regulating corticotrope function. Altogether, these results provide compelling evidence that CORT plays a key, sex-dependent role in the regulation of the GH/ACTH axes in response to fasting.
Collapse
Affiliation(s)
- José Cordoba-Chacón
- Maimonides Institute of Biomedical Research of Cordoba (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); Department of Cell Biology, Physiology, and Immunology (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), University of Córdoba; Hospital Universitario Reina Sofía (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); and Campus de Excelencia Internacional Agroalimentario (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Córdoba 14004, Spain; Department of Medicine (J.C.-C.), Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago and Jesse Brown Veteran Affairs Medical Center, Research and Development Division, Chicago, Illinois 60612; and Department of Psychiatry and Behavioral Sciences (L.d.L.), Stanford University School of Medicine, Palo Alto, California 94305
| | - Manuel D Gahete
- Maimonides Institute of Biomedical Research of Cordoba (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); Department of Cell Biology, Physiology, and Immunology (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), University of Córdoba; Hospital Universitario Reina Sofía (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); and Campus de Excelencia Internacional Agroalimentario (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Córdoba 14004, Spain; Department of Medicine (J.C.-C.), Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago and Jesse Brown Veteran Affairs Medical Center, Research and Development Division, Chicago, Illinois 60612; and Department of Psychiatry and Behavioral Sciences (L.d.L.), Stanford University School of Medicine, Palo Alto, California 94305
| | - Ana I Pozo-Salas
- Maimonides Institute of Biomedical Research of Cordoba (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); Department of Cell Biology, Physiology, and Immunology (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), University of Córdoba; Hospital Universitario Reina Sofía (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); and Campus de Excelencia Internacional Agroalimentario (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Córdoba 14004, Spain; Department of Medicine (J.C.-C.), Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago and Jesse Brown Veteran Affairs Medical Center, Research and Development Division, Chicago, Illinois 60612; and Department of Psychiatry and Behavioral Sciences (L.d.L.), Stanford University School of Medicine, Palo Alto, California 94305
| | - Luis de Lecea
- Maimonides Institute of Biomedical Research of Cordoba (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); Department of Cell Biology, Physiology, and Immunology (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), University of Córdoba; Hospital Universitario Reina Sofía (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); and Campus de Excelencia Internacional Agroalimentario (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Córdoba 14004, Spain; Department of Medicine (J.C.-C.), Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago and Jesse Brown Veteran Affairs Medical Center, Research and Development Division, Chicago, Illinois 60612; and Department of Psychiatry and Behavioral Sciences (L.d.L.), Stanford University School of Medicine, Palo Alto, California 94305
| | - Justo P Castaño
- Maimonides Institute of Biomedical Research of Cordoba (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); Department of Cell Biology, Physiology, and Immunology (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), University of Córdoba; Hospital Universitario Reina Sofía (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); and Campus de Excelencia Internacional Agroalimentario (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Córdoba 14004, Spain; Department of Medicine (J.C.-C.), Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago and Jesse Brown Veteran Affairs Medical Center, Research and Development Division, Chicago, Illinois 60612; and Department of Psychiatry and Behavioral Sciences (L.d.L.), Stanford University School of Medicine, Palo Alto, California 94305
| | - Raúl M Luque
- Maimonides Institute of Biomedical Research of Cordoba (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); Department of Cell Biology, Physiology, and Immunology (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), University of Córdoba; Hospital Universitario Reina Sofía (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.); and Campus de Excelencia Internacional Agroalimentario (J.C.-C., M.D.G., A.I.P.-S., J.P.C., R.M.L.), Córdoba 14004, Spain; Department of Medicine (J.C.-C.), Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago and Jesse Brown Veteran Affairs Medical Center, Research and Development Division, Chicago, Illinois 60612; and Department of Psychiatry and Behavioral Sciences (L.d.L.), Stanford University School of Medicine, Palo Alto, California 94305
| |
Collapse
|
16
|
Lack of cortistatin or somatostatin differentially influences DMBA-induced mammary gland tumorigenesis in mice in an obesity-dependent mode. Breast Cancer Res 2016; 18:29. [PMID: 26956474 PMCID: PMC4782371 DOI: 10.1186/s13058-016-0689-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/19/2016] [Indexed: 01/08/2023] Open
Abstract
Background Somatostatin (SST) and cortistatin (CORT), two structurally and functionally related peptides, share a family of widespread receptors (sst1-5) to exert apparently similar biological actions, including endocrine/metabolic regulation and suppression of tumor cell proliferation. However, despite their therapeutic potential, attempts to apply SST-analogs to treat breast cancer have yielded unsatisfactory results. Actually, the specific roles of SST and CORT in mammary gland tumorigenesis (MGT), particularly in relation to metabolic dysregulation (i.e. obesity), remain unknown. Methods The role of endogenous SST and CORT in carcinogen-induced MGT was investigated under normal (lean) and obesity conditions. To that end, SST- and CORT-knockout (KO) mice and their respective littermate-controls, fed low-fat (LF) or high-fat (HF) diets, were treated with 7,12-dimethyl-benza-anthracene (DMBA) once a week (wk) for 3 wk, and MGT was monitored for 25 wk. Additionally, we examined the effect of SST or CORT removal in the development of the mammary gland. Results Lack of SST did not alter DMBA-induced MGT incidence under lean conditions; conversely, lack of endogenous CORT severely aggravated DMBA-induced MGT in LF-fed mice. These differences were not attributable to altered mammary gland development. HF-diet modestly increased the sensitivity to DMBA-induced carcinogenesis in control mice, whereas, as observed in LF-fed CORT-KO, HF-fed CORT-KO mice exhibited aggravated tumor incidence, discarding a major influence of obesity on these CORT actions. In marked contrast, HF-fed SST-KO mice exhibited much higher tumor incidence than LF-fed SST-KO mice, which could be associated with higher mammary complexity. Conclusions Endogenous SST and CORT distinctly impact on DMBA-induced MGT, in a manner that is strongly dependent on the metabolic/endocrine milieu (lean vs. obese status). Importantly, CORT, rather than SST, could represent a major inhibitor of MGT under normal/lean-conditions, whereas both neuropeptides would similarly influence MGT under obesity conditions. The mechanisms mediating these different effects likely involve mammary development and hormones, but the precise underlying factors are still to be fully elucidated. However, our findings comprise suggestive evidence that CORT-like molecules, rather than classic SST-analogs, may help to identify novel tools for the medical treatment of breast cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0689-1) contains supplementary material, which is available to authorized users.
Collapse
|
17
|
Tremblay M, Brisson D, Gaudet D. Association between a polymorphic poly-T repeat sequence in the promoter of the somatostatin gene and hypertension. Hypertens Res 2016; 39:467-74. [PMID: 26818653 DOI: 10.1038/hr.2016.4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/07/2015] [Accepted: 12/19/2015] [Indexed: 02/04/2023]
Abstract
Despite the numerous common pathways connecting blood pressure regulation to somatostatin (SST) metabolism, the SST gene has never been seen as a significant blood pressure modulator. The aim of this study was to evaluate the association between a poly-T repeat sequence (rs34872250) in the promoter of the SST gene and blood pressure, according to the obesity status. We genotyped 1918 French-Canadian subjects from a founder population. Analyses were performed according to the length of the poly-T repeat sequence on both alleles and divided into two groups, the 13/13-13/14 group and the 13/15-13/16 group. The effect of age, gender, body mass index, antihypertensive drugs and diabetic status were considered. Systolic, diastolic and mean blood pressures are significantly higher among the 13/15-13/16 group in the whole sample (P<0.05). Whereas the differences remain significant in women, they turn to be non-significant when men are considered alone. The risk of hypertension is increased in the 13/15-13/16 group, particularly among overweight/obese subjects. Systolic blood pressure is significantly higher among overweight/obese carriers of the 13/15-13/16 alleles in the whole sample (P<0.001), in men (P=0.006) and in women (P=0.002), even after correction for age and antihypertensive drugs. These results suggest that the poly-T repeat sequence polymorphism in the promoter of the SST gene is associated with significant variations of blood pressure and could modulate the risk of hypertension, particularly among women.
Collapse
Affiliation(s)
- Monique Tremblay
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal, Chicoutimi, Québec, Canada.,ECOGENE-21, Clinical and Translational Research Center, Chicoutimi, Québec, Canada
| | - Diane Brisson
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal, Chicoutimi, Québec, Canada.,ECOGENE-21, Clinical and Translational Research Center, Chicoutimi, Québec, Canada
| | - Daniel Gaudet
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal, Chicoutimi, Québec, Canada.,ECOGENE-21, Clinical and Translational Research Center, Chicoutimi, Québec, Canada
| |
Collapse
|
18
|
Pedraza-Arévalo S, Córdoba-Chacón J, Pozo-Salas AI, L-López F, de Lecea L, Gahete MD, Castaño JP, Luque RM. Not So Giants: Mice Lacking Both Somatostatin and Cortistatin Have High GH Levels but Show No Changes in Growth Rate or IGF-1 Levels. Endocrinology 2015; 156:1958-64. [PMID: 25830706 DOI: 10.1210/en.2015-1132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Somatostatin (SST) and cortistatin (CORT) are two highly related neuropeptides involved in the regulation of various endocrine secretions. In particular, SST and CORT are two primary negative regulators of GH secretion. Consequently, single SST or CORT knockout mice exhibit elevated GH levels; however, this does not lead to increased IGF-1 levels or somatic growth. This apparent lack of correspondence has been suggested to result from compensatory mechanisms between both peptides. To test this hypothesis, in this study we explored, for the first time, the consequences of simultaneously deleting endogenous SST and CORT by generating a double SST/CORT knockout mouse model and exploring its endocrine and metabolic phenotype. Our results demonstrate that simultaneous deletion of SST and CORT induced a drastic elevation of endogenous GH levels, which, surprisingly, did not lead to changes in growth rate or IGF-1 levels, suggesting the existence of additional factors/systems that, in the absence of endogenous SST and CORT, could counteract GH actions. Notably, elevation in circulating GH levels were not accompanied by changes in pituitary GH expression or by alterations in the expression of its main regulators (GHRH and ghrelin) or their receptors (GHRH receptor, GHS receptor, or SST/CORT receptors) at the hypothalamic or pituitary level. However, although double-SST/CORT knockout male mice exhibited normal glucose and insulin levels, they had improved insulin sensitivity compared with the control mice. Therefore, these results suggest the existence of an intricate interplay among the known (SST/CORT), and likely unknown, inhibitory components of the GH/IGF-1 axis to regulate somatic growth and glucose/insulin homeostasis.
Collapse
Affiliation(s)
- S Pedraza-Arévalo
- Department of Cell Biology, Physiology, and Immunology (S.P.-A., J.C.-C., A.I.P.-S., F.L.L., M.D.G., J.P.C., R.M.L.), University of Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, and Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), E-14014 Córdoba, Spain; Section of Endocrinology, Diabetes, and Metabolism (J.C.-C.), Department of Medicine (J.C.-C.), University of Illinois at Chicago, Chicago, Illinois 60637; Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612; and Department of Psychiatry and Behavioral Sciences (L.d.L.), Stanford University, Stanford, California 94305
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Luque RM, Córdoba-Chacón J, Ibáñez-Costa A, Gesmundo I, Grande C, Gracia-Navarro F, Tena-Sempere M, Ghigo E, Gahete MD, Granata R, Kineman RD, Castaño JP. Obestatin plays an opposite role in the regulation of pituitary somatotrope and corticotrope function in female primates and male/female mice. Endocrinology 2014; 155:1407-17. [PMID: 24484169 PMCID: PMC3959609 DOI: 10.1210/en.2013-1728] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Obestatin is a 23-amino-acid amidated peptide that is encoded by the ghrelin gene. Previous studies have shown obestatin can modulate the hypothalamic neuronal circuitry that regulates pituitary function, perhaps by modulating the actions of ghrelin. However, the direct actions of obestatin on pituitary function remain controversial. Here, primary pituitary cell cultures from a nonhuman primate (baboon) and mice were used to test the effects of obestatin on pituitary hormone expression and secretion. In pituitary cultures from both species, obestatin had no effect on prolactin, LH, FSH, or TSH expression/release. Conversely, obestatin stimulated proopiomelanocortin expression and ACTH release and inhibited GH expression/release in vitro, actions that were also observed in vivo in mice treated with obestatin. In vitro, obestatin inhibited the stimulatory actions of ghrelin on GH but not ACTH release. The inhibitory effect of obestatin on somatotrope function was associated with an overall reduction in pituitary transcription factor-1 and GHRH receptor mRNA levels in vitro and in vivo as well as a reduction in hypothalamic GHRH and ghrelin expression in vivo. The stimulatory effect of obestatin on ACTH was associated with an increase in pituitary CRF receptors. Obestatin also reduced the expression of pituitary somatostatin receptors (sst1/sst2), which could serve to modify its impact on hormone secretion. The in vitro actions of obestatin on both GH and ACTH release required the adenylyl cyclase and MAPK routes. Taken together, our results provide evidence that obestatin can act directly at the pituitary to control somatotrope and corticotrope function, and these effects are conserved across species.
Collapse
Affiliation(s)
- Raúl M Luque
- Department of Cell Biology, Physiology, and Immunology (R.M.L., J.C.-C., A.I.-C., F.G.-N., M.T.-S., M.D.G., J.P.C.), University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofia, and Centros de Investigación Biomédica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, E-14014 Córdoba, Spain; Department of Medicine (J.C.-C., R.D.K.), University of Illinois at Chicago, and Jesse Brown Veterans Affairs Medical Center (J.C.-C., R.D.K.), Research and Development Division, Chicago, Illinois 60612; and Department of Medical Sciences (I.G., C.G., E.G., R.G.), University of Torino, 10126 Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Álvarez-Crespo M, Martínez-Sánchez N, Ruíz-Pino F, Garcia-Lavandeira M, Alvarez CV, Tena-Sempere M, Nogueiras R, Diéguez C, López M. The orexigenic effect of orexin-A revisited: dependence of an intact growth hormone axis. Endocrinology 2013; 154:3589-98. [PMID: 23861376 DOI: 10.1210/en.2013-1251] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Fifteen years ago orexins were identified as central regulators of energy homeostasis. Since then, that concept has evolved considerably and orexins are currently considered, besides orexigenic neuropeptides, key modulators of sleep-wake cycle and neuroendocrine function. Little is known, however, about the effect of the neuroendocrine milieu on orexins' effects on energy balance. We therefore investigated whether hypothalamic-pituitary axes have a role in the central orexigenic action of orexin A (OX-A) by centrally injecting hypophysectomized, adrenalectomized, gonadectomized (male and female), hypothyroid, and GH-deficient dwarf rats with OX-A. Our data showed that the orexigenic effect of OX-A is fully maintained in adrenalectomized and gonadectomized (females and males) rats, slightly reduced in hypothyroid rats, and totally abolished in hypophysectomized and dwarf rats when compared with their respective vehicle-treated controls. Of note, loss of the OX-A effect on feeding was associated with a blunted OX-A-induced increase in the expression of either neuropeptide Y or its putative regulator, the transcription factor cAMP response-element binding protein, as well as its phosphorylated form, in the arcuate nucleus of the hypothalamus of hypophysectomized and dwarf rats. Overall, this evidence suggests that the orexigenic action of OX-A depends on an intact GH axis and that this neuroendocrine feedback loop may be of interest in the understanding of orexins action on energy balance and GH deficiency.
Collapse
Affiliation(s)
- Mayte Álvarez-Crespo
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain and CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Córdoba-Chacón J, Gahete MD, Pozo-Salas AI, Castaño JP, Kineman RD, Luque RM. Endogenous somatostatin is critical in regulating the acute effects of L-arginine on growth hormone and insulin release in mice. Endocrinology 2013; 154:2393-8. [PMID: 23696563 PMCID: PMC3689276 DOI: 10.1210/en.2013-1136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
l-arginine (l-Arg) rapidly stimulates GH and insulin release in vivo. It has been hypothesized that l-Arg stimulates GH release by lowering hypothalamic somatostatin (SST) tone. l-Arg may also act directly at the pituitary to stimulate GH release. Moreover, l-Arg has a direct stimulatory effect on β-cells, which is thought to be blunted by the release of SST from pancreatic δ-cells. To confirm the role of endogenous SST on l-Arg-induced GH and insulin release, wild-type (WT) and SST-knockout (SST-KO) mice were injected with l-Arg (ip; 0.8 g/kg), and pre-/post-injection GH, insulin, and glucose levels were measured. In WT mice, l-Arg evoked a 6-fold increase in circulating GH. However, there was only a modest increase in GH levels in WT pituitary cell cultures treated with l-Arg. In contrast, l-Arg failed to increase GH in SST-KO beyond their already elevated levels. These results further support the hypothesis that the primary mechanism by which l-Arg acutely increases GH in vivo is by lowering hypothalamic SST input to the pituitary and not via direct pituitary effects. Additionally, l-Arg induced a clear first-phase insulin secretion in WT mice, but not in SST-KO. However, SST-KO, but not WT mice, displayed a robust and sustained second-phase insulin release. These results further support a role for endogenous SST in regulating l-Arg-mediated insulin release.
Collapse
|
22
|
Gahete MD, Córdoba-Chacón J, Lin Q, Brüning JC, Kahn CR, Castaño JP, Christian H, Luque RM, Kineman RD. Insulin and IGF-I inhibit GH synthesis and release in vitro and in vivo by separate mechanisms. Endocrinology 2013; 154:2410-20. [PMID: 23671263 PMCID: PMC3689283 DOI: 10.1210/en.2013-1261] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
IGF-I is considered a primary inhibitor of GH secretion. Insulin may also play an important role in regulating GH levels because insulin, like IGF-I, can suppress GH synthesis and release in primary pituitary cell cultures and insulin is negatively correlated with GH levels in vivo. However, understanding the relative contribution insulin and IGF-I exert on controlling GH secretion has been hampered by the fact that circulating insulin and IGF-I are regulated in parallel and insulin (INSR) and IGF-I (IGFIR) receptors are structurally/functionally related and ubiquitously expressed. To evaluate the separate roles of insulin and IGF-I in directly regulating GH secretion, we used the Cre/loxP system to knock down the INSR and IGFIR in primary mouse pituitary cell cultures and found insulin-mediated suppression of GH is independent of the IGFIR. In addition, pharmacological blockade of intracellular signals in both mouse and baboon cultures revealed insulin requires different pathways from IGF-I to exert a maximal inhibitory effect on GH expression/release. In vivo, somatotrope-specific knockout of INSR (SIRKO) or IGFIR (SIGFRKO) increased GH levels. However, comparison of the pattern of GH release, GH expression, somatotrope morphometry, and pituitary explant sensitivity to acute GHRH challenge in lean SIRKO and SIGFRKO mice strongly suggests the primary role of insulin in vivo is to suppress GH release, whereas IGF-I serves to regulate GH synthesis. Finally, SIRKO and/or SIGFRKO could not prevent high-fat, diet-induced suppression of pituitary GH expression, indicating other factors/tissues are involved in the decline of GH observed with weight gain.
Collapse
Affiliation(s)
- Manuel D Gahete
- Research and Development Division, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Oberg K, Casanovas O, Castaño JP, Chung D, Delle Fave G, Denèfle P, Harris P, Khan MS, Kulke MH, Scarpa A, Tang LH, Wiedenmann B. Molecular pathogenesis of neuroendocrine tumors: implications for current and future therapeutic approaches. Clin Cancer Res 2013; 19:2842-9. [PMID: 23459719 DOI: 10.1158/1078-0432.ccr-12-3458] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The treatment landscape and biologic understanding of neuroendocrine tumors (NET) has shifted dramatically in recent years. Recent studies have shown that somatostatin analogues have the potential not only to control symptoms of hormone hypersecretion but also have the ability to slow tumor growth in patients with advanced carcinoid. The results of clinical trials have further shown that the VEGF pathway inhibitor sunitinib and the mTOR inhibitor everolimus have efficacy in patients with advanced pancreatic NETs. The efficacy of these targeted therapies in NET suggests that the molecular characterization of NETs may provide an avenue to predict both which patients may benefit most from the treatment and to overcome potential drug resistance. Recent genomic studies of NETs have further suggested that pathways regulating chromatin remodeling and epigenetic modification may play a key role in regulating NET growth. These observations offer the potential for new therapeutic and diagnostic advances for patients with NET.
Collapse
Affiliation(s)
- Kjell Oberg
- Department of Endocrine Oncology, University Hospital, Uppsala, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW To describe our current understanding of the mechanisms involved in the regulation of linear growth in childhood obesity. RECENT FINDINGS The developmental origins hypothesis has focused on low birth weight individuals with subsequent obesity, identifying a cascade of neuroendocrine regulatory factors involved in the progressive increase in body fat and metabolic risk. Yet, tall stature is the common clinical outcome of childhood obesity. Recent data have expanded our understanding of environmental influences on developing systems. Here, we review the elements of neuroendocrine systems contributing to the integration of metabolic controls involved in growth regulation in the obese child with particular emphasis on growth hormone, ghrelin, insulin-like growth factors and insulin. SUMMARY Growth patterns of obesity during childhood are well described, documenting increased linear growth in early childhood associated with accelerated pubertal maturation resulting in normal adult height. Despite recent data suggesting that ghrelin and the growth hormone secretagogue receptor, as well as the insulin-like growth factors, their binding proteins and insulin have potential to be mediators of nutrient exposure and linear growth, it remains to be determined how these systems interrelate and determine growth. This is an area of ongoing investigation.
Collapse
Affiliation(s)
- Ilene Fennoy
- Division of Pediatric Endocrinology, Diabetes and Metabolism, Department of Pediatrics, Columbia University Medical Center, New York, New York, USA.
| |
Collapse
|
25
|
Gruszka A, Culler MD, Melmed S. Somatostatin analogs and chimeric somatostatin-dopamine molecules differentially regulate human growth hormone and prolactin gene expression and secretion in vitro. Mol Cell Endocrinol 2012; 362:104-9. [PMID: 22705877 DOI: 10.1016/j.mce.2012.05.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 12/21/2011] [Accepted: 05/31/2012] [Indexed: 01/31/2023]
Abstract
We tested effects of selective somatostatin receptor 2 (SST2) agonist BIM-23120, SST5 agonist BIM-23206 and chimeric somatostatin-dopamine molecules (SRIF/DA) BIM-23A760 and BIM-23A761 on GH and PRL secretion and gene expression in human GH/PRL-secreting pituitary tumors in vitro. In "responders" group BIM-23120 suppressed GH levels by 26±4%, BIM-23206 by 31±5%, BIM-23A760 by 23±4%, BIM-23A761 by 39±8% and D(2)-dopamine agonist BIM-53097 by 31±5%. Using real-time PCR we demonstrated that GH inhibition was not accompanied by decreased GH mRNA levels. PRL secretion was inhibited by BIM-23A760 (29±5%), BIM-23A761 (34±4%), BIM-23206 (26±4%) and BIM-53097 (36±2%). SRIF/DA and BIM-53097 also suppressed PRL mRNA levels. Concluding, SST2 and SST5 agonists and SRIF/DA inhibit GH secretion, but do not suppress GH gene transcription. SRIF/DA and BIM-53097 inhibit both PRL secretion and PRL gene expression. SST5 agonist inhibits PRL secretion, but does not suppress PRL gene expression. D(2) affinity is crucial in SRIF/DA action on PRL gene expression.
Collapse
Affiliation(s)
- Anna Gruszka
- Division of Endocrinology, Cedars-Sinai Research Institute, University of California School of Medicine, Los Angeles, CA 90048, USA.
| | | | | |
Collapse
|
26
|
Samaco RC, McGraw CM, Ward CS, Sun Y, Neul JL, Zoghbi HY. Female Mecp2(+/-) mice display robust behavioral deficits on two different genetic backgrounds providing a framework for pre-clinical studies. Hum Mol Genet 2012; 22:96-109. [PMID: 23026749 DOI: 10.1093/hmg/dds406] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Rett syndrome (RTT) is an X-linked neurological disorder caused by mutations in the gene encoding the transcriptional modulator methyl-CpG-binding protein 2 (MeCP2). Typical RTT primarily affects girls and is characterized by a brief period of apparently normal development followed by the loss of purposeful hand skills and language, the onset of anxiety, hand stereotypies, autistic features, seizures and autonomic dysfunction. Mecp2 mouse models have extensively been studied to demonstrate the functional link between MeCP2 dysfunction and RTT pathogenesis. However, the majority of studies have focused primarily on the molecular and behavioral consequences of the complete absence of MeCP2 in male mice. Studies of female Mecp2(+/-) mice have been limited because of potential phenotypic variability due to X chromosome inactivation effects. To determine whether reproducible and reliable phenotypes can be detected Mecp2(+/-) mice, we analyzed Mecp2(+/-) mice of two different F1 hybrid isogenic backgrounds and at young and old ages using several neurobehavioral and physiological assays. Here, we report a multitude of phenotypes in female Mecp2(+/-) mice, some presenting as early as 5 weeks of life. We demonstrate that Mecp2(+/-) mice recapitulate several aspects of typical RTT and show that mosaic expression of MeCP2 does not preclude the use of female mice in behavioral and molecular studies. Importantly, we uncover several behavioral abnormalities that are present in two genetic backgrounds and report on phenotypes that are unique to one background. These findings provide a framework for pre-clinical studies aimed at improving the constellation of phenotypes in a mouse model of RTT.
Collapse
Affiliation(s)
- Rodney C Samaco
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
27
|
Córdoba-Chacón J, Gahete MD, Culler MD, Castaño JP, Kineman RD, Luque RM. Somatostatin dramatically stimulates growth hormone release from primate somatotrophs acting at low doses via somatostatin receptor 5 and cyclic AMP. J Neuroendocrinol 2012; 24:453-63. [PMID: 22129035 DOI: 10.1111/j.1365-2826.2011.02261.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Somatostatin and cortistatin have been shown to act directly on pituitary somatotrophs to inhibit growth hormone (GH) release. However, previous results from nonprimate species indicate that these peptides can also directly stimulate GH secretion, at low concentrations. The relevance of this phenomenon in a nonhuman primate model was investigated in the present study by testing the impact of somatostatin/cortistatin on GH release in primary pituitary cell cultures from baboons. High doses (> 10(-10) m) of somatostatin/cortistatin did not alter basal GH secretion but blocked GH-releasing hormone (GHRH)- and ghrelin-induced GH release. However, at low concentrations (10(-17)-10(-13) m), somatostatin/cortistatin dramatically stimulated GH release to levels comparable to those evoked by GHRH or ghrelin. Use of somatostatin receptor (sst) specific agonists/antagonists, and signal transduction blockers indicated that sst2 and sst1 activation via intact adenylate cylcase and mitogen-activated protein kinase systems mediated the inhibitory actions of high-concentration somatostatin. By contrast, the stimulatory actions of low-dose somatostatin on GH release were mediated by sst5 signalling through adenylate cylcase/cAMP/protein kinase A and intracellular Ca(2+) pathways, and were additive with ghrelin (not GHRH). Notably, low-concentrations of somatostatin, similar to sst5-agonists, inhibited prolactin release. These results clearly demonstrate that the ultimate impact of somatostatin/cortistatin on hormone release is dose-dependent, cell type-selective and receptor-specific, where the stimulatory effects of low-concentration somatostatin/cortistatin on GH release extend to primates, thereby supporting the notion that this action is relevant in regulating GH secretion in humans.
Collapse
Affiliation(s)
- J Córdoba-Chacón
- Department of Cell Biology, Physiology and Immunology, University of Cordoba and Reina Sofia University Hospital, Instituto Maimónides de Investigación Biomédica de Córdoba, and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
| | | | | | | | | | | |
Collapse
|
28
|
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: 1.1] [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
|
29
|
Dobolyi A, Dimitrov E, Palkovits M, Usdin TB. The neuroendocrine functions of the parathyroid hormone 2 receptor. Front Endocrinol (Lausanne) 2012; 3:121. [PMID: 23060860 PMCID: PMC3465808 DOI: 10.3389/fendo.2012.00121] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 09/20/2012] [Indexed: 01/25/2023] Open
Abstract
The G-protein coupled parathyroid hormone 2 receptor (PTH2R) is concentrated in endocrine and limbic regions in the forebrain. Its endogenous ligand, tuberoinfundibular peptide of 39 residues (TIP39), is synthesized in only two brain regions, within the posterior thalamus and the lateral pons. TIP39-expressing neurons have a widespread projection pattern, which matches the PTH2R distribution in the brain. Neuroendocrine centers including the preoptic area, the periventricular, paraventricular, and arcuate nuclei contain the highest density of PTH2R-positive networks. The administration of TIP39 and an antagonist of the PTH2R as well as the investigation of mice that lack functional TIP39 and PTH2R revealed the involvement of the PTH2R in a variety of neural and neuroendocrine functions. TIP39 acting via the PTH2R modulates several aspects of the stress response. It evokes corticosterone release by activating corticotropin-releasing hormone-containing neurons in the hypothalamic paraventricular nucleus. Block of TIP39 signaling elevates the anxiety state of animals and their fear response, and increases stress-induced analgesia. TIP39 has also been suggested to affect the release of additional pituitary hormones including arginine-vasopressin and growth hormone. A role of the TIP39-PTH2R system in thermoregulation was also identified. TIP39 may play a role in maintaining body temperature in a cold environment via descending excitatory pathways from the preoptic area. Anatomical and functional studies also implicated the TIP39-PTH2R system in nociceptive information processing. Finally, TIP39 induced in postpartum dams may play a role in the release of prolactin during lactation. Potential mechanisms leading to the activation of TIP39 neurons and how they influence the neuroendocrine system are also described. The unique TIP39-PTH2R neuromodulator system provides the possibility for developing drugs with a novel mechanism of action to control neuroendocrine disorders.
Collapse
Affiliation(s)
- Arpád Dobolyi
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Hungarian Academy of Sciences, Semmelweis UniversityBudapest, Hungary
- *Correspondence: Arpád Dobolyi, Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Hungarian Academy of Sciences, Semmelweis University, Tűzoltó u. 58, Budapest H-1094, Hungary. e-mail:
| | - Eugene Dimitrov
- Section on Fundamental Neuroscience, National Institute of Mental Health, National Institute of HealthBethesda, MD, USA
| | - Miklós Palkovits
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Hungarian Academy of Sciences, Semmelweis UniversityBudapest, Hungary
| | - Ted B. Usdin
- Section on Fundamental Neuroscience, National Institute of Mental Health, National Institute of HealthBethesda, MD, USA
| |
Collapse
|
30
|
Córdoba-Chacón J, Gahete MD, Pozo-Salas AI, Martínez-Fuentes AJ, de Lecea L, Gracia-Navarro F, Kineman RD, Castaño JP, Luque RM. Cortistatin is not a somatostatin analogue but stimulates prolactin release and inhibits GH and ACTH in a gender-dependent fashion: potential role of ghrelin. Endocrinology 2011; 152:4800-12. [PMID: 21971153 PMCID: PMC3230064 DOI: 10.1210/en.2011-1542] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Cortistatin (CST) and somatostatin (SST) evolve from a common ancestral gene and share remarkable structural, pharmacological, and functional homologies. Although CST has been considered as a natural SST-analogue acting through their shared receptors (SST receptors 1-5), emerging evidence indicates that these peptides might in fact exert unique roles via selective receptors [e.g. CST, not SST, binds ghrelin receptor growth hormone secretagogue receptor type 1a (GHS-R1a)]. To determine whether the role of endogenous CST is different from SST, we characterized the endocrine-metabolic phenotype of male/female CST null mice (cort-/-) at hypothalamic-pituitary-systemic (pancreas-stomach-adrenal-liver) levels. Also, CST effects on hormone expression/secretion were evaluated in primary pituitary cell cultures from male/female mice and female primates (baboons). Specifically, CST exerted an unexpected stimulatory role on prolactin (PRL) secretion, because both male/female cort-/- mice had reduced PRL levels, and CST treatment (in vivo and in vitro) increased PRL secretion, which could be blocked by a GHS-R1a antagonist in vitro and likely relates to the decreased success of female cort-/- in first-litter pup care at weaning. In contrast, CST inhibited GH and adrenocorticotropin-hormone axes in a gender-dependent fashion. In addition, a rise in acylated ghrelin levels was observed in female cort-/- mice, which were associated with an increase in stomach ghrelin/ghrelin O-acyl transferase expression. Finally, CST deficit uncovered a gender-dependent role of this peptide in the regulation of glucose-insulin homeostasis, because male, but not female, cort-/- mice developed insulin resistance. The fact that these actions are not mimicked by SST and are strongly gender dependent offers new grounds to investigate the hitherto underestimated physiological relevance of CST in the regulation of physiological/metabolic processes.
Collapse
|
31
|
Córdoba-Chacón J, Gahete MD, Castaño JP, Kineman RD, Luque RM. Somatostatin and its receptors contribute in a tissue-specific manner to the sex-dependent metabolic (fed/fasting) control of growth hormone axis in mice. Am J Physiol Endocrinol Metab 2011; 300:E46-54. [PMID: 20943754 PMCID: PMC3023207 DOI: 10.1152/ajpendo.00514.2010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Somatostatin (SST) inhibits growth hormone (GH) secretion and regulates multiple processes by signaling through its receptors sst1-5. Differential expression of SST/ssts may contribute to sex-specific GH pattern and fasting-induced GH rise. To further delineate the tissue-specific roles of SST and sst1-5 in these processes, their expression patterns were evaluated in hypothalamus, pituitary, and stomach of male and female mice under fed/fasted conditions in the presence (wild type) or absence (SST-knockout) of endogenous SST. Under fed conditions, hypothalamic/stomach SST/ssts expression did not differ between sexes, whereas male pituitary expressed more SST and sst2A/2B/3/5A/5TMD2/5TMD1 and less sst1, and male pituitary cell cultures were more responsive to SST inhibitory actions on GH release compared with females. This suggests that local pituitary SST/ssts can contribute to the sexually dimorphic pattern of GH release. Fasting (48 h) reduced stomach sst2A/B and hypothalamic SST/sst2A expression in both sexes, whereas it caused a generalized downregulation of pituitary sst subtypes in male and of sst2A only in females. Thus, fasting can reduce SST sensitivity across tissues and SST input to the pituitary, thereby jointly contributing to enhance GH release. In SST-knockout mice, lack of SST differentially altered sst subtype expression levels in both sexes, supporting an important role for SST in sex-dependent control of GH axis. Evaluation of SST, IGF-I, and glucocorticoid effects on hypothalamic and pituitary cell cultures revealed that these hormones could directly account for alterations in sst2/5 expression in the physiological states examined. Taken together, these results indicate that changes in SST output and sensitivity can contribute critically to precisely define, in a tissue-dependent manner, the sex-specific metabolic regulation of the GH axis.
Collapse
Affiliation(s)
- José Córdoba-Chacón
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba, and Centro de Investigacion Biomedica en Red Fisiopatología de la Obesidad y Nutrición, Cordoba, Spain
| | | | | | | | | |
Collapse
|
32
|
Taboada GF, Neto LV, Luque RM, Córdoba-Chacón J, de Oliveira Machado E, de Carvalho DP, Kineman RD, Gadelha MR. Impact of gsp oncogene on the mRNA content for somatostatin and dopamine receptors in human somatotropinomas. Neuroendocrinology 2011; 93:40-7. [PMID: 21079388 DOI: 10.1159/000322040] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 10/13/2010] [Indexed: 12/27/2022]
Abstract
INTRODUCTION It has been reported in some series that gsp+ somatotropinomas are more sensitive to somatostatin analogues (SA) and dopamine's actions which may be related to their somatostatin receptor (SSTR) and dopamine receptor (DR) profile. No previous studies have been undertaken to evaluate the SSTR and DR profile related with the gsp status in somatotropinomas. OBJECTIVES To determine if (1) gsp status is correlated with response to octreotide LAR (LAR) and tumor expression patterns of SSTR1-5 and DR1-5 and (2) cAMP level can directly modulate SSTR and DR mRNA levels. METHODS Response to SA was evaluated by GH and IGF-I percent reduction after 3 and 6 months of treatment with LAR. Conventional PCR and sequencing were used to identify gsp+ tumors. Quantitative real-time PCR was used to determine SSTR and DR tumor expression. Primary pituitary cell cultures of primates were used to study whether SSTR and DR expression is regulated by forskolin. RESULTS The response to LAR did not significantly differ between patients with gsp+ and gsp- tumors; however, gsp+ tumors expressed higher levels of SSTR1, SSTR2, DR2 and a lower level of SSTR3. Forskolin increased SSTR1, SSTR2, DR1 and DR2 expression in cell cultures. CONCLUSION Elevated SSTR1, SSTR2, and DR2 tumor expression may help improve responsiveness to SA and DA therapy; however, this study may not have been appropriately powered to observe significant effects in the clinical response. Elevated cAMP levels could be directly responsible for the upregulation in SSTR1, SSTR2 and DR2 mRNA levels observed in gsp+ patients.
Collapse
|
33
|
Richalet JP, Letournel M, Souberbielle JC. Effects of high-altitude hypoxia on the hormonal response to hypothalamic factors. Am J Physiol Regul Integr Comp Physiol 2010; 299:R1685-92. [PMID: 20926759 DOI: 10.1152/ajpregu.00484.2010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acute and chronic exposure to high altitude induces various physiological changes, including activation or inhibition of various hormonal systems. In response to activation processes, a desensitization of several pathways has been described, especially in the adrenergic system. In the present study, we aimed to assess whether the hypophyseal hormones are also subjected to a hypoxia-induced decrease in their response to hypothalamic factors. Basal levels of hormones and the responses of TSH, thyroid hormones, prolactin, sex hormones, and growth hormone to the injection of TRH, gonadotropin-releasing hormone, and growth hormone-releasing hormone (GHRH) were studied in eight men in normoxia and on prolonged exposure (3-4 days) to an altitude of 4,350 m. Thyroid hormones were elevated at altitude (+16 to +21%), while TSH levels were unchanged, and follicle-stimulating hormone and prolactin decreased, while leutinizing hormone was unchanged. Norepinephrine and cortisol levels were elevated, while no change was observed in levels of epinephrine, dopamine, growth hormone (GH), IGF-1, and IGFBP-3. The mean response to hypothalamic factors was similar in both altitudes for all studied hormones, although total T4 was lower in hypoxia during 45 to 60 min after injection. The effect of hypoxia on the hypophyseal response to hypothalamic factors was similar among subjects, except for the GH response to GHRH administration. We conclude that prolonged exposure to high-altitude hypoxia induces contrasted changes in hormonal levels, but the hypophyseal response to hypothalamic factors does not appear to be blunted.
Collapse
Affiliation(s)
- Jean-Paul Richalet
- Université Paris 13, Unité de Formation et de Recherche Santé Médecine Biologie Humaine, Laboratoire Réponses Cellulaires et Fonctionnelles à l'Hypoxie, Bobigny, France.
| | | | | |
Collapse
|
34
|
Sheridan MA, Hagemeister AL. Somatostatin and somatostatin receptors in fish growth. Gen Comp Endocrinol 2010; 167:360-5. [PMID: 19735661 DOI: 10.1016/j.ygcen.2009.09.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 08/20/2009] [Accepted: 09/02/2009] [Indexed: 01/25/2023]
Abstract
Multiple forms of somatostatin (SS) and SS receptors (SSTR) are produced widely in the tissues of fish and interact to coordinate numerous physiological processes. Insight into their role in growth regulation emerged from studies of abnormal growth and of whole animals. The influence of SS on organismal growth operates at several levels of the growth hormone (GH)-insulin-like growth factor-1 (IGF-1) system. SS inhibits production and release of pituitary GH, but not all forms of SS are equipotent in this action. SS also influences the GH-IGF-1 system in an extrapituitary manner by reducing sensitivity to GH as well as by inhibiting IGF-1 production and secretion, and diminishing IGF-1 sensitivity. Peripheral actions of SS are important for the local control of growth and may help to coordinate growth with other processes such as metabolism, development, and reproduction by reprogramming cell responsiveness.
Collapse
Affiliation(s)
- Mark A Sheridan
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58108-6050, USA.
| | | |
Collapse
|
35
|
Gahete MD, Córdoba-Chacón J, Salvatori R, Castaño JP, Kineman RD, Luque RM. Metabolic regulation of ghrelin O-acyl transferase (GOAT) expression in the mouse hypothalamus, pituitary, and stomach. Mol Cell Endocrinol 2010; 317:154-60. [PMID: 20035826 PMCID: PMC2819060 DOI: 10.1016/j.mce.2009.12.023] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 12/16/2009] [Indexed: 01/07/2023]
Abstract
Ghrelin acts as an endocrine link connecting physiological processes regulating food intake, body composition, growth, and energy balance. Ghrelin is the only peptide known to undergo octanoylation. The enzyme mediating this process, ghrelin O-acyltransferase (GOAT), is expressed in the gastrointestinal tract (GI; primary source of circulating ghrelin) as well as other tissues. The present study demonstrates that stomach GOAT mRNA levels correlate with circulating acylated-ghrelin levels in fasted and diet-induced obese mice. In addition, GOAT was found to be expressed in both the pituitary and hypothalamus (two target tissues of ghrelin's actions), and regulated in response to metabolic status. Using primary pituitary cell cultures as a model system to study the regulation of GOAT expression, we found that acylated-ghrelin, but not desacyl-ghrelin, increased GOAT expression. In addition, growth-hormone-releasing hormone (GHRH) and leptin increased, while somatostatin (SST) decreased GOAT expression. The physiologic relevance of these later results is supported by the observation that pituitary GOAT expression in mice lacking GHRH, SST and leptin showed opposite changes to those observed after in vitro treatment with the corresponding peptides. Therefore, it seems plausible that these hormones directly contribute to the regulation of pituitary GOAT. Interestingly, in all the models studied, pituitary GOAT expression paralleled changes in the expression of a dominant spliced-variant of ghrelin (In2-ghrelin) and therefore this transcript may be a primary substrate for pituitary GOAT. Collectively, these observations support the notion that the GI tract is not the only source of acylated-ghrelin, but in fact locally produced des-acylated-ghrelin could be converted to acylated-ghrelin within target tissues by locally active GOAT, to mediate its tissue-specific effects.
Collapse
Affiliation(s)
- Manuel D. Gahete
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, 14004 Córdoba, Spain
| | - Jose Córdoba-Chacón
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, 14004 Córdoba, Spain
| | | | - Justo P. Castaño
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, 14004 Córdoba, Spain
| | - Rhonda D. Kineman
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, University of Illinois at Chicago, Chicago, IL, 60612
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL, 60612
| | - Raul M. Luque
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición, 14004 Córdoba, Spain
| |
Collapse
|
36
|
Córdoba-Chacón J, Gahete MD, Duran-Prado M, Pozo-Salas AI, Malagón MM, Gracia-Navarro F, Kineman RD, Luque RM, Castaño JP. Identification and characterization of new functional truncated variants of somatostatin receptor subtype 5 in rodents. Cell Mol Life Sci 2010; 67:1147-63. [PMID: 20063038 PMCID: PMC11115927 DOI: 10.1007/s00018-009-0240-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 12/08/2009] [Accepted: 12/18/2009] [Indexed: 12/11/2022]
Abstract
Somatostatin and cortistatin exert multiple biological actions through five receptors (sst1-5); however, not all their effects can be explained by activation of sst1-5. Indeed, we recently identified novel truncated but functional human sst5-variants, present in normal and tumoral tissues. In this study, we identified and characterized three novel truncated sst5 variants in mice and one in rats displaying different numbers of transmembrane-domains [TMD; sst5TMD4, sst5TMD2, sst5TMD1 (mouse-variants) and sst5TMD1 (rat-variant)]. These sst5 variants: (1) are functional to mediate ligand-selective-induced variations in [Ca(2+)]i and cAMP despite being truncated; (2) display preferential intracellular distribution; (3) mostly share full-length sst5 tissue distribution, but exhibit unique differences; (4) are differentially regulated by changes in hormonal/metabolic environment in a tissue- (e.g., central vs. systemic) and ligand-dependent manner. Altogether, our results demonstrate the existence of new truncated sst5-variants with unique ligand-selective signaling properties, which could contribute to further understanding the complex, distinct pathophysiological roles of somatostatin and cortistatin.
Collapse
Affiliation(s)
- Jose Córdoba-Chacón
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Edificio Severo Ochoa. Planta 3. Campus de Rabanales, 14014 Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn 06/03), Córdoba, Spain
| | - Manuel D. Gahete
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Edificio Severo Ochoa. Planta 3. Campus de Rabanales, 14014 Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn 06/03), Córdoba, Spain
| | - Mario Duran-Prado
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Edificio Severo Ochoa. Planta 3. Campus de Rabanales, 14014 Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn 06/03), Córdoba, Spain
| | - Ana I. Pozo-Salas
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Edificio Severo Ochoa. Planta 3. Campus de Rabanales, 14014 Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn 06/03), Córdoba, Spain
| | - María M. Malagón
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Edificio Severo Ochoa. Planta 3. Campus de Rabanales, 14014 Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn 06/03), Córdoba, Spain
| | - F. Gracia-Navarro
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Edificio Severo Ochoa. Planta 3. Campus de Rabanales, 14014 Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn 06/03), Córdoba, Spain
| | - Rhonda D. Kineman
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL USA
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL USA
| | - Raul M. Luque
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Edificio Severo Ochoa. Planta 3. Campus de Rabanales, 14014 Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn 06/03), Córdoba, Spain
| | - Justo P. Castaño
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Edificio Severo Ochoa. Planta 3. Campus de Rabanales, 14014 Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn 06/03), Córdoba, Spain
| |
Collapse
|
37
|
Ben-Shlomo A, Melmed S. Pituitary somatostatin receptor signaling. Trends Endocrinol Metab 2010; 21:123-33. [PMID: 20149677 PMCID: PMC2834886 DOI: 10.1016/j.tem.2009.12.003] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 12/09/2009] [Accepted: 12/10/2009] [Indexed: 12/27/2022]
Abstract
Somatotropin-release inhibitory factor (SRIF) is a major regulator of pituitary function, mostly inhibiting hormone secretion and to a lesser extent pituitary cell growth. Five SRIF receptor subtypes (SSTR1-5) are ubiquitously expressed G-protein coupled receptors. In the pituitary, SSTR1, 2, 3 and 5 are expressed, with SSTR2 and SSTR5 predominating. As new SRIF analogs have recently been introduced for treatment of pituitary disease, we evaluate the current knowledge of cell-specific pituitary SRIF receptor signaling and highlight areas of future research for comprehensive understanding of these mechanisms. Elucidating pituitary SRIF receptor signaling enables understanding of pituitary hormone secretion and cell growth, and also encourages future therapeutic development for pituitary disorders.
Collapse
Affiliation(s)
- Anat Ben-Shlomo
- Pituitary Center, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California 90048, USA
| | | |
Collapse
|
38
|
Ben-Shlomo A, Zhou C, Pichurin O, Chesnokova V, Liu NA, Culler MD, Melmed S. Constitutive somatostatin receptor activity determines tonic pituitary cell response. Mol Endocrinol 2009; 23:337-48. [PMID: 19131507 DOI: 10.1210/me.2008-0361] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Somatostatin (SRIF) binds G protein-coupled SRIF receptor subtypes (SST1, -2, -3, -4, and -5) to regulate cell secretion and proliferation. Hypothalamic SRIF inhibits pituitary growth hormone, thyroid stimulating hormone, and ACTH secretion. We tested SRIF-independent constitutive SST activity in AtT20 mouse pituitary corticotroph cells in which ACTH secretion is highly sensitive to SRIF action. Stable transfectants expressing SST2 or SST5 were sensitized to selective agonist action, and constitutive SST receptor activity was demonstrated by forskolin and pertussis toxin cAMP cell responses. Persistent constitutive SST activity decreased cell ACTH responses to CRH through decreased expression of CRH receptor subtype 1. Decreased dopamine receptor type 1 expression was associated with attenuated dopamine agonist action, whereas responses to isoproterenol were enhanced through increased beta2-adrenoreceptor expression. Thus, integrated pituitary cell ACTH regulation is determined both by phasic SRIF action, as well as by tonic constitutive SST activity, independently of SRIF.
Collapse
Affiliation(s)
- Anat Ben-Shlomo
- Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Academic Affairs, Room 2015, Los Angeles, California 90048, USA
| | | | | | | | | | | | | |
Collapse
|