1
|
Kineman RD, Del Rio-Moreno M, Waxman DJ. Liver-specific actions of GH and IGF1 that protect against MASLD. Nat Rev Endocrinol 2025; 21:105-117. [PMID: 39322791 DOI: 10.1038/s41574-024-01037-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; also known as nonalcoholic fatty liver disease) is a chronic condition associated with metabolic syndrome, a group of conditions that includes obesity, insulin resistance, hyperlipidaemia and cardiovascular disease. Primary growth hormone (GH) deficiency is associated with MASLD, and the decline in circulating levels of GH with weight gain might contribute to the development of MASLD. Raising endogenous GH secretion or administering GH replacement therapy in the context of MASLD enhances insulin-like growth factor 1 (IGF1) production and reduces steatosis and the severity of liver injury. GH and IGF1 indirectly control MASLD progression by regulating systemic metabolic function. Evidence supports the proposal that GH and IGF1 also have a direct role in regulating liver metabolism and health. This Review focuses on how GH acts on the hepatocyte in a sex-dependent manner to limit lipid accumulation, reduce stress, and promote survival and regeneration. In addition, we discuss how GH and IGF1 might regulate non-parenchymal cells of the liver to control inflammation and fibrosis, which have a major effect on hepatocyte survival and regeneration. Development of a better understanding of how GH and IGF1 coordinate the functions of specific, individual liver cell types might provide insight into the aetiology of MASLD initiation and progression and suggest novel approaches for the treatment of MASLD.
Collapse
Affiliation(s)
- Rhonda D Kineman
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA.
| | - Mercedes Del Rio-Moreno
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
2
|
Pérez-Gómez JM, Montero-Hidalgo AJ, Luque RM. GHRH and reproductive systems: Mechanisms, functions, and clinical implications. Rev Endocr Metab Disord 2024:10.1007/s11154-024-09931-8. [PMID: 39612161 DOI: 10.1007/s11154-024-09931-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 11/30/2024]
Abstract
Growth hormone-releasing hormone (GHRH) has classically been considered a regulatory neuropeptide of the hypothalamic-pituitary system, which mediates its anabolic effects through hepatic GH/IGF-I axis. However, during the last decades it has been demonstrated that this key regulatory hormone may be produced in numerous peripheral tissues outside the central nervous system, participating in fundamental physiological functions through a complex balance between its purely endocrine action, and the recently local (autocrine/paracrine) discovered role. Among peripheral sites, its presence in the male and female reproductive systems stands out. In this review, we will first explore the role of the GHRH/GHRH-R hormone axis as a central player in the gonadal function; then, we will discuss available information regarding the presence of GHRH/GHRH-R and the potential physiological roles in reproductive systems of various species; and finally, we will address how reproductive system-related disorders-such as infertility problems, endometriosis, or tumor pathologies (including prostate, or ovarian cancer)-could benefit from hormonal interventions related to the manipulation of the GHRH axis.
Collapse
Affiliation(s)
- Jesús M Pérez-Gómez
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), IMIBIC Building. Av. Menéndez Pidal S/N. 14004, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
| | - Antonio J Montero-Hidalgo
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), IMIBIC Building. Av. Menéndez Pidal S/N. 14004, 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), IMIBIC Building. Av. Menéndez Pidal S/N. 14004, 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, (CIBERobn), Cordoba, Spain.
| |
Collapse
|
3
|
Voltan G, Mazzeo P, Regazzo D, Scaroni C, Ceccato F. Role of Estrogen and Estrogen Receptor in GH-Secreting Adenomas. Int J Mol Sci 2023; 24:9920. [PMID: 37373068 DOI: 10.3390/ijms24129920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/17/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Acromegaly is a rare disease with several systemic complications that may lead to increased overall morbidity and mortality. Despite several available treatments, ranging from transsphenoidal resection of GH-producing adenomas to different medical therapies, complete hormonal control is not achieved in some cases. Some decades ago, estrogens were first used to treat acromegaly, resulting in a significant decrease in IGF1 levels. However, due to the consequent side effects of the high dose utilized, this treatment was later abandoned. The evidence that estrogens are able to blunt GH activity also derives from the evidence that women with GH deficiency taking oral estro-progestins pills need higher doses of GH replacement therapy. In recent years, the role of estrogens and Selective Estrogens Receptor Modulators (SERMs) in acromegaly treatment has been re-evaluated, especially considering poor control of the disease under first- and second-line medical treatment. In this review, we analyze the state of the art concerning the impact of estrogen and SERMs on the GH/IGF1 axis, focusing on molecular pathways and the possible implications for acromegaly treatment.
Collapse
Affiliation(s)
- Giacomo Voltan
- Department of Medicine (DIMED), University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Endocrinology Unit, Padova University Hospital, Via Ospedale Civile 105, 35128 Padova, Italy
| | - Pierluigi Mazzeo
- Department of Medicine (DIMED), University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Endocrinology Unit, Padova University Hospital, Via Ospedale Civile 105, 35128 Padova, Italy
| | - Daniela Regazzo
- Department of Medicine (DIMED), University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Carla Scaroni
- Department of Medicine (DIMED), University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Endocrinology Unit, Padova University Hospital, Via Ospedale Civile 105, 35128 Padova, Italy
| | - Filippo Ceccato
- Department of Medicine (DIMED), University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Endocrinology Unit, Padova University Hospital, Via Ospedale Civile 105, 35128 Padova, Italy
| |
Collapse
|
4
|
Mirczuk SM, Scudder CJ, Read JE, Crossley VJ, Regan JT, Richardson KM, Simbi B, McArdle CA, Church DB, Fenn J, Kenny PJ, Volk HA, Wheeler-Jones CP, Korbonits M, Niessen SJ, McGonnell IM, Fowkes RC. Natriuretic Peptide Expression and Function in GH3 Somatolactotropes and Feline Somatotrope Pituitary Tumours. Int J Mol Sci 2021; 22:ijms22031076. [PMID: 33499110 PMCID: PMC7865297 DOI: 10.3390/ijms22031076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 11/17/2022] Open
Abstract
Patients harbouring mutations in genes encoding C-type natriuretic peptide (CNP; NPPC) or its receptor guanylyl cyclase B (GC-B, NPR2) suffer from severe growth phenotypes; loss-of-function mutations cause achondroplasia, whereas gain-of-function mutations cause skeletal overgrowth. Although most of the effects of CNP/GC-B on growth are mediated directly on bone, evidence suggests the natriuretic peptides may also affect anterior pituitary control of growth. Our previous studies described the expression of NPPC and NPR2 in a range of human pituitary tumours, normal human pituitary, and normal fetal human pituitary. However, the natriuretic peptide system in somatotropes has not been extensively explored. Here, we examine the expression and function of the CNP/GC-B system in rat GH3 somatolactotrope cell line and pituitary tumours from a cohort of feline hypersomatotropism (HST; acromegaly) patients. Using multiplex RT-qPCR, all three natriuretic peptides and their receptors were detected in GH3 cells. The expression of Nppc was significantly enhanced following treatment with either 100 nM TRH or 10 µM forskolin, yet only Npr1 expression was sensitive to forskolin stimulation; the effects of forskolin and TRH on Nppc expression were PKA- and MAPK-dependent, respectively. CNP stimulation of GH3 somatolactotropes significantly inhibited Esr1, Insr and Lepr expression, but dramatically enhanced cFos expression at the same time point. Oestrogen treatment significantly enhanced expression of Nppa, Nppc, Npr1, and Npr2 in GH3 somatolactotropes, but inhibited CNP-stimulated cGMP accumulation. Finally, transcripts for all three natriuretic peptides and receptors were expressed in feline pituitary tumours from patients with HST. NPPC expression was negatively correlated with pituitary tumour volume and SSTR5 expression, but positively correlated with D2R and GHR expression. Collectively, these data provide mechanisms that control expression and function of CNP in somatolactotrope cells, and identify putative transcriptional targets for CNP action in somatotropes.
Collapse
Affiliation(s)
- Samantha M. Mirczuk
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (S.M.M.); (C.J.S.); (J.E.R.); (V.J.C.); (J.T.R.); (K.M.R.)
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (B.S.); (C.P.W.-J.); (I.M.M.)
| | - Christopher J. Scudder
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (S.M.M.); (C.J.S.); (J.E.R.); (V.J.C.); (J.T.R.); (K.M.R.)
- Clinical Sciences & Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (D.B.C.); (J.F.); (P.J.K.); (H.A.V.); (S.J.N.)
| | - Jordan E. Read
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (S.M.M.); (C.J.S.); (J.E.R.); (V.J.C.); (J.T.R.); (K.M.R.)
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (B.S.); (C.P.W.-J.); (I.M.M.)
| | - Victoria J. Crossley
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (S.M.M.); (C.J.S.); (J.E.R.); (V.J.C.); (J.T.R.); (K.M.R.)
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (B.S.); (C.P.W.-J.); (I.M.M.)
| | - Jacob T. Regan
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (S.M.M.); (C.J.S.); (J.E.R.); (V.J.C.); (J.T.R.); (K.M.R.)
| | - Karen M. Richardson
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (S.M.M.); (C.J.S.); (J.E.R.); (V.J.C.); (J.T.R.); (K.M.R.)
| | - Bigboy Simbi
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (B.S.); (C.P.W.-J.); (I.M.M.)
| | - Craig A. McArdle
- Department of Translational Science, Bristol Medical School, University of Bristol, Whitson Street, Bristol BS1 3NY, UK;
| | - David B. Church
- Clinical Sciences & Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (D.B.C.); (J.F.); (P.J.K.); (H.A.V.); (S.J.N.)
| | - Joseph Fenn
- Clinical Sciences & Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (D.B.C.); (J.F.); (P.J.K.); (H.A.V.); (S.J.N.)
| | - Patrick J. Kenny
- Clinical Sciences & Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (D.B.C.); (J.F.); (P.J.K.); (H.A.V.); (S.J.N.)
- Small Animal Specialist Hospital, 1 Richardson Place, North Ryde, 2113 NSW, Australia
| | - Holger A. Volk
- Clinical Sciences & Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (D.B.C.); (J.F.); (P.J.K.); (H.A.V.); (S.J.N.)
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Bünteweg 9, 30559 Hannover, Germany
| | - Caroline P. Wheeler-Jones
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (B.S.); (C.P.W.-J.); (I.M.M.)
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK;
| | - Stijn J. Niessen
- Clinical Sciences & Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (D.B.C.); (J.F.); (P.J.K.); (H.A.V.); (S.J.N.)
| | - Imelda M. McGonnell
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (B.S.); (C.P.W.-J.); (I.M.M.)
| | - Robert C. Fowkes
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (S.M.M.); (C.J.S.); (J.E.R.); (V.J.C.); (J.T.R.); (K.M.R.)
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (B.S.); (C.P.W.-J.); (I.M.M.)
- Correspondence: ; Tel.: +11-44-207-468-1215
| |
Collapse
|
5
|
Garcia-Galiano D, Cara AL, Tata Z, Allen SJ, Myers MG, Schipani E, Elias CF. ERα Signaling in GHRH/Kiss1 Dual-Phenotype Neurons Plays Sex-Specific Roles in Growth and Puberty. J Neurosci 2020; 40:9455-9466. [PMID: 33158965 PMCID: PMC7724138 DOI: 10.1523/jneurosci.2069-20.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/07/2020] [Accepted: 10/25/2020] [Indexed: 02/07/2023] Open
Abstract
Gonadal steroids modulate growth hormone (GH) secretion and the pubertal growth spurt via undefined central pathways. GH-releasing hormone (GHRH) neurons express estrogen receptor α (ERα) and androgen receptor (AR), suggesting changing levels of gonadal steroids during puberty directly modulate the somatotropic axis. We generated mice with deletion of ERα in GHRH cells (GHRHΔERα), which displayed reduced body length in both sexes. Timing of puberty onset was similar in both groups, but puberty completion was delayed in GHRHΔERα females. Lack of AR in GHRH cells (GHRHΔAR mice) induced no changes in body length, but puberty completion was also delayed in females. Using a mouse model with two reporter genes, we observed that, while GHRHtdTom neurons minimally colocalize with Kiss1hrGFP in prepubertal mice, ∼30% of GHRH neurons coexpressed both reporter genes in adult females, but not in males. Developmental analysis of Ghrh and Kiss1 expression suggested that a subpopulation of ERα neurons in the arcuate nucleus of female mice undergoes a shift in phenotype, from GHRH to Kiss1, during pubertal transition. Our findings demonstrate that direct actions of gonadal steroids in GHRH neurons modulate growth and puberty and indicate that GHRH/Kiss1 dual-phenotype neurons play a sex-specific role in the crosstalk between the somatotropic and gonadotropic axes during pubertal transition.SIGNIFICANCE STATEMENT Late maturing adolescents usually show delayed growth and bone age. At puberty, gonadal steroids have stimulatory effects on the activation of growth and reproductive axes, but the existence of gonadal steroid-sensitive neuronal crosstalk remains undefined. Moreover, the neural basis for the sex differences observed in the clinical arena is unknown. Lack of ERα in GHRH neurons disrupts growth in both sexes and causes pubertal delay in females. Deletion of androgen receptor in GHRH neurons only delayed female puberty. In adult females, not males, a subset of GHRH neurons shift phenotype to start producing Kiss1. Thus, direct estrogen action in GHRH/Kiss1 dual-phenotype neurons modulates growth and puberty and may orchestrate the sex differences in endocrine function observed during pubertal transition.
Collapse
Affiliation(s)
| | | | - Zachary Tata
- Department of Orthopedic Surgery, Medicine, and Cell and Developmental Biology
| | | | - Martin G Myers
- Department of Molecular and Integrative Physiology
- Department of Internal Medicine Division of Metabolism, Endocrinology and Diabetes
| | - Ernestina Schipani
- Department of Orthopedic Surgery, Medicine, and Cell and Developmental Biology
| | - Carol F Elias
- Department of Molecular and Integrative Physiology
- Department of Gynecology and Obstetrics, University of Michigan, Ann Arbor, Michigan 48109-5622
| |
Collapse
|
6
|
Birzniece V, McLean M, Reddy N, Ho KKY. Disparate Effect of Aromatization on the Central Regulation of GH Secretion by Estrogens in Men and Postmenopausal Women. J Clin Endocrinol Metab 2019; 104:2978-2984. [PMID: 30920620 DOI: 10.1210/jc.2019-00265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/22/2019] [Indexed: 01/28/2023]
Abstract
CONTEXT Estrogen receptor antagonism by tamoxifen inhibits GH secretion in both men and postmenopausal women, suggesting that estrogen, albeit at low concentration, stimulates GH secretion. However, systemic estrogen replacement in postmenopausal women does not enhance GH secretion. To clarify the role of estrogen in mediating GH secretion, we investigated the effect of estrogen deprivation by using aromatase inhibitors. AIM To determine whether estrogens mediate GH secretion in men and postmenopausal women. DESIGN The effects of letrozole, an aromatase inhibitor, and tamoxifen were compared in an open-label crossover study. Eight men and 14 women received tamoxifen (20 mg/d) and letrozole (2.5 mg/d) for 2 weeks each. The primary endpoints were GH response to arginine stimulation and gonadal steroid levels. RESULTS In men, letrozole significantly (P < 0.05) reduced the peak GH response to arginine (mean ± SEM; Δ -49.4% ± 18.1%). Tamoxifen also reduced the mean peak GH, but this did not reach statistical significance. In postmenopausal women, letrozole did not affect peak GH, whereas tamoxifen significantly (P < 0.05) reduced peak GH (Δ -47.3% ± 10%). In men, letrozole reduced circulating estradiol (from 43.1 ± 2.8 to 12.7 ± 1.3 pmol/L; P < 0.001), whereas in women estradiol was undetectable (<11 pmol/L) at baseline and throughout letrozole therapy. CONCLUSION Because estrogen deprivation reduced circulating GH, we conclude that estrogens regulate GH secretion in men. In postmenopausal women, the neutral effect of aromatase inhibition is likely explained by pre-existing estrogen deficiency. The inhibition of GH secretion by tamoxifen in menopause suggests a non-estrogen receptor-mediated mechanism of action. In contrast to men, estrogen is unlikely to mediate GH secretion in postmenopausal women.
Collapse
Affiliation(s)
- Vita Birzniece
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Medicine, Western Sydney University, Penrith, New South Wales, Australia
- Department of Diabetes and Endocrinology, Blacktown Hospital, Blacktown, New South Wales, Australia
- School of Medicine, University of New South Wales, New South Wales, Australia
| | - Mark McLean
- School of Medicine, Western Sydney University, Penrith, New South Wales, Australia
| | - Navneeta Reddy
- Department of Diabetes and Endocrinology, Blacktown Hospital, Blacktown, New South Wales, Australia
| | - Ken K Y Ho
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Roelfsema F, Yang RJ, Takahashi PY, Erickson D, Bowers CY, Veldhuis JD. Aromatized Estrogens Amplify Nocturnal Growth Hormone Secretion in Testosterone-Replaced Older Hypogonadal Men. J Clin Endocrinol Metab 2018; 103:4419-4427. [PMID: 30032193 PMCID: PMC6212797 DOI: 10.1210/jc.2018-00755] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/20/2018] [Indexed: 11/19/2022]
Abstract
CONTEXT Testosterone (T) increases GH secretion in older men with a relative lack of T, in hypogonadal men of all ages, and in patients undergoing sex reassignment. The role of estradiol (E2) in men is less well defined. OBJECTIVE To assess the contribution of aromatization of T to spontaneous nocturnal and stimulated GH secretion. PARTICIPANTS Four groups of healthy older men (N = 74, age range 57 to 77 years) were studied. The gonadotropic axis was clamped with the gonadotropin-releasing hormone antagonist degarelix. Three groups received T and one group placebo addback. Two T-replaced groups were treated with anastrozole (an aromatase inhibitor) and either placebo or E2 addback. MAIN OUTCOME MEASURES Ten-minute GH concentration profiles were quantified by deconvolution analysis, after overnight (2200 to 0800 hours) sampling, and after combined IV injection of GHRH (0.3 µg/kg) and GHRH-2 (0.3 µg/kg) and withdrawal of a 2-hour somatostatin infusion (1 µg/kg/h). RESULTS E2 addback during aromatase inhibition increased basal (P = 0.046), pulsatile (P = 0.020), and total (P = 0.018) GH secretion by 60% to 70%. E2 did not potentiate GH secretory stimuli. Logarithmically transformed pulsatile GH secretion correlated strongly and positively with concurrent E2 concentrations overall (P = 0.028) and under anastrozole treatment (P = 0.005). CONCLUSION E2 administration in older men transdermally stimulates overnight pulsatile GH secretion. The exact site of E2 action cannot be ascertained from these experiments but may include hypothalamic loci involved in GH regulation, especially because GH secretagogue effects on somatotrope pituitary cells were not affected.
Collapse
Affiliation(s)
- Ferdinand Roelfsema
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, Netherlands
| | - Rebecca J Yang
- Endocrine Research Unit, Mayo School of Graduate Medical Education, Center for Translational Science Activities, Mayo Clinic, Rochester, Minnesota
| | - Paul Y Takahashi
- Department of Primary Care Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Dana Erickson
- Division of Endocrinology, Mayo Clinic, Rochester, Minnesota
| | - Cyril Y Bowers
- Department of Internal Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Johannes D Veldhuis
- Endocrine Research Unit, Mayo School of Graduate Medical Education, Center for Translational Science Activities, Mayo Clinic, Rochester, Minnesota
- Correspondence and Reprint Requests: Johannes D. Veldhuis, MD, Endocrine Research Unit, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905. E-mail:
| |
Collapse
|
8
|
Impact of aromatase absence on murine intraocular pressure and retinal ganglion cells. Sci Rep 2018; 8:3280. [PMID: 29459742 PMCID: PMC5818491 DOI: 10.1038/s41598-018-21475-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/31/2018] [Indexed: 12/14/2022] Open
Abstract
We hypothesize that aromatase, an enzyme that regulates estrogen production, plays a significant role in the control of intraocular pressure (IOP) and retinal ganglion cells (RGCs). To begin to test our hypothesis, we examined the impact of aromatase absence, which completely eliminates estrogen synthesis, in male and female mice. Studies were performed with adult, age-matched wild type (WT) and aromatase knockout (ArKO) mice. IOP was measured in a masked fashion in both eyes of conscious mice at 12 and 24 weeks of age. Retinas were obtained and processed for RGC counting with a confocal microscope. IOP levels in both 12- and 24-week old female ArKO mice were significantly higher than those of age- and sex-matched WT controls. The mean increase in IOP was 7.9% in the 12-week-, and 19.7% in the 24-week-old mice, respectively. These changes were accompanied by significant 9% and 7% decreases in RGC numbers in the ArKO female mice, relative to controls, at 12- and 24-weeks, respectively. In contrast, aromatase deficiency did not lead to an increased IOP in male mice. There was a significant reduction in RGC counts in the 12-, but not 24-, week-old male ArKO mice, as compared to their age- and sex-matched WT controls. Overall, our findings show that aromatase inhibition in females is associated with elevated IOP and reduced RGC counts.
Collapse
|
9
|
Roelfsema F, Yang RJ, Takahashi PY, Erickson D, Bowers CY, Veldhuis JD. Effects of Toremifene, a Selective Estrogen Receptor Modulator, on Spontaneous and Stimulated GH Secretion, IGF-I, and IGF-Binding Proteins in Healthy Elderly Subjects. J Endocr Soc 2017; 2:154-165. [PMID: 29383334 PMCID: PMC5789038 DOI: 10.1210/js.2017-00457] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 12/21/2017] [Indexed: 11/19/2022] Open
Abstract
Context: Estrogens amplify spontaneous and stimulated growth hormone (GH) secretion, whereas they diminish GH-dependent insulin-like growth factor (IGF)-I in a dose-dependent manner. Selective estrogen receptor modulators (SERMs), including tamoxifen and toremifene, are widely adjunctively used in breast and prostate cancer. Although some endocrine effects of tamoxifen are known, few data are available for toremifene. Objective: To explore sex-dependent effects of toremifene on spontaneous 10-hour overnight GH secretion, followed by GH-releasing hormone–ghrelin stimulation. Additionally, effects on IGF-I, its binding proteins, and sex hormone–binding globulin (SHBG) were quantified. Participants and Design: Twenty men and 20 women, within an allowable age range of 50 to 80 years, volunteered for this double-blind, placebo-controlled prospective crossover study. Ten-minute blood sampling was done for 10 hours overnight and then for 2 hours after combined GH-releasing hormone–ghrelin injection. Main Outcome Measures: Pulsatile GH and stimulated GH secretion, and fasting levels of IGF-I, IGF-binding protein (IGFBP)1, IGFBP3, and SHBG. Results: Toremifene did not enhance pulsatile or stimulated GH secretion, but decreased IGF-I by 20% in men and women. IGFBP3 was unchanged, whereas while IGFBP1 and SHBG increased in both sexes to a similar extent. Conclusions: The expected rise in spontaneous and stimulated GH secretion under the diminished negative feedback restraint of powered IGF-I favors a central inhibitory antiestrogenic effect of toremifene. Estrogenic effects of toremifene on the liver were present, as evidenced by increased IGFBP1 and SHBG levels. Men and women responded to this SERM comparably.
Collapse
Affiliation(s)
- Ferdinand Roelfsema
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Rebecca J Yang
- Endocrine Research Unit, Mayo School of Graduate Medical Education, Center for Translational Science Activities, Mayo Clinic, Rochester, Minnesota 55905
| | - Paul Y Takahashi
- Department of Primary Care Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Dana Erickson
- Department of Endocrinology, Mayo Clinic, Rochester, Minnesota 55905
| | - Cyril Y Bowers
- Department of Internal Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana 70112
| | - Johannes D Veldhuis
- Endocrine Research Unit, Mayo School of Graduate Medical Education, Center for Translational Science Activities, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
10
|
Birzniece V, Ho KKY. Sex steroids and the GH axis: Implications for the management of hypopituitarism. Best Pract Res Clin Endocrinol Metab 2017; 31:59-69. [PMID: 28477733 DOI: 10.1016/j.beem.2017.03.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Growth hormone (GH) regulates somatic growth, substrate metabolism and body composition. Sex hormones exert profound effect on the secretion and action of GH. Estrogens stimulate the secretion of GH, but inhibit the action of GH on the liver, an effect that occurs when administered orally. Estrogens suppress GH receptor signaling by stimulating the expression proteins that inhibit cytokine receptor signaling. This effect of estrogens is avoided when physiological doses of estrogens are administered via a non-oral route. Estrogen-like compounds, such as selective estrogen receptor modulators, possess dual properties of inhibiting the secretion as well as the action of GH. In contrast, androgens stimulate GH secretion, driving IGF-1 production. In the periphery, androgens enhance the action of GH. The differential effects of estrogens and androgens influence the dose of GH replacement in patients with hypopituitarism on concomitant treatment with sex steroids. Where possible, a non-oral route of estrogen replacement is recommended for optimizing cost-benefit of GH replacement in women with GH deficiency. Adequate androgen replacement in conjunction with GH replacement is required to achieve the full anabolic effect in men with hypopituitarism.
Collapse
Affiliation(s)
- Vita Birzniece
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; School of Medicine, University of New South Wales, NSW 2052, Australia.
| | - Ken K Y Ho
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; Centres for Health Research, Princess Alexandra Hospital, University of Queensland, Brisbane, QLD 4102, Australia
| |
Collapse
|
11
|
Bentz AB, Sirman AE, Wada H, Navara KJ, Hood WR. Relationship between maternal environment and DNA methylation patterns of estrogen receptor alpha in wild Eastern Bluebird (Sialia sialis) nestlings: a pilot study. Ecol Evol 2016; 6:4741-52. [PMID: 27547309 PMCID: PMC4979703 DOI: 10.1002/ece3.2162] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 12/18/2022] Open
Abstract
There is mounting evidence that, across taxa, females breeding in competitive environments tend to allocate more testosterone to their offspring prenatally and these offspring typically have more aggressive and faster‐growing phenotypes. To date, no study has determined the mechanisms mediating this maternal effect's influence on offspring phenotype. However, levels of estrogen receptor alpha (ERα) gene expression are linked to differences in early growth and aggression; thus, maternal hormones may alter gene regulation, perhaps via DNA methylation, of ERα in offspring during prenatal development. We performed a pilot study to examine natural variation in testosterone allocation to offspring through egg yolks in wild Eastern Bluebirds (Sialia sialis) in varying breeding densities and percent DNA methylation of CG dinucleotides in the ERα promoter in offspring brain regions associated with growth and behavior. We hypothesized that breeding density would be positively correlated with yolk testosterone, and prenatal exposure to maternal‐derived yolk testosterone would be associated with greater offspring growth and decreased ERα promoter methylation. Yolk testosterone concentration was positively correlated with breeding density, nestling growth rate, and percent DNA methylation of one out of five investigated CpG sites (site 3) in the diencephalon ERα promoter, but none in the telencephalon (n = 10). Percent DNA methylation of diencephalon CpG site 3 was positively correlated with growth rate. These data suggest a possible role for epigenetics in mediating the effects of the maternal environment on offspring phenotype. Experimentally examining this mechanism with a larger sample size in future studies may help elucidate a prominent way in which animals respond to their environment. Further, by determining the mechanisms that mediate maternal effects, we can begin to understand the potential for the heritability of these mechanisms and the impact that maternal effects are capable of producing at an evolutionary scale.
Collapse
Affiliation(s)
- Alexandra B Bentz
- Poultry Science Department University of Georgia 203 Poultry Science Bldg. Athens Georigia 30602
| | - Aubrey E Sirman
- Department of Biological Sciences Auburn University 101 Life Science Building Auburn Alabama 36849
| | - Haruka Wada
- Department of Biological Sciences Auburn University 101 Life Science Building Auburn Alabama 36849
| | - Kristen J Navara
- Poultry Science Department University of Georgia 203 Poultry Science Bldg. Athens Georigia 30602
| | - Wendy R Hood
- Department of Biological Sciences Auburn University 101 Life Science Building Auburn Alabama 36849
| |
Collapse
|
12
|
Birzniece V, Ho KKY. Estrogen receptor antagonism uncovers gender-dimorphic suppression of whole body fat oxidation in humans: differential effects of tamoxifen on the GH and gonadal axes. Eur J Endocrinol 2015. [PMID: 26199431 DOI: 10.1530/eje-15-0426] [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: 12/13/2022]
Abstract
CONTEXT Tamoxifen, a selective estrogen receptor modulator, suppresses GH secretion in women but not in men. It increases testosterone levels in men. As GH and testosterone stimulate fat metabolism, the metabolic consequences of tamoxifen may be greater in women than in men. OBJECTIVE To determine whether tamoxifen suppresses fat oxidation (Fox) to a greater degree in women than in men. DESIGN An open-label study of ten healthy postmenopausal women and ten healthy men receiving 2-week treatment with tamoxifen (20 mg/day). ENDPOINT MEASURES GH response to arginine stimulation, serum levels of IGF1, testosterone and LH (men only), sex hormone binding globulin (SHBG) and whole body basal and postprandial Fox. RESULTS In women, tamoxifen significantly reduced the mean GH response to arginine stimulation (Δ -87%, P<0.05) and circulating IGF1 levels (Δ -23.5±5.4%, P<0.01). Tamoxifen reduced postprandial Fox in women (Δ -34.6±10.3%; P<0.05). In men, tamoxifen did not affect the GH response to arginine stimulation but significantly reduced mean IGF1 levels (Δ -24.8±6.1%, P<0.01). Tamoxifen increased mean testosterone levels (Δ 52±14.2%; P<0.01). Fox was not significantly affected by tamoxifen in men. CONCLUSION Tamoxifen attenuated the GH response to stimulation and reduced postprandial Fox in women but not in men. We conclude that at a therapeutic dose, the suppressive effect of tamoxifen on fat metabolism is gender-dependent. Higher testosterone levels may mitigate the suppression of GH secretion and Fox during tamoxifen treatment in men.
Collapse
Affiliation(s)
- Vita Birzniece
- Department of EndocrinologyGarvan Institute of Medical Research, St Vincent's Hospital, Sydney, New South Wales 2010, AustraliaSchool of MedicineUniversity of Western Sydney, Sydney, New South Wales 2148, AustraliaThe University of NSWSydney, New South Wales 2052, AustraliaCentres for Health ResearchPrincess Alexandra Hospital, The University of Queensland, Brisbane, Queensland 4102, Australia Department of EndocrinologyGarvan Institute of Medical Research, St Vincent's Hospital, Sydney, New South Wales 2010, AustraliaSchool of MedicineUniversity of Western Sydney, Sydney, New South Wales 2148, AustraliaThe University of NSWSydney, New South Wales 2052, AustraliaCentres for Health ResearchPrincess Alexandra Hospital, The University of Queensland, Brisbane, Queensland 4102, Australia Department of EndocrinologyGarvan Institute of Medical Research, St Vincent's Hospital, Sydney, New South Wales 2010, AustraliaSchool of MedicineUniversity of Western Sydney, Sydney, New South Wales 2148, AustraliaThe University of NSWSydney, New South Wales 2052, AustraliaCentres for Health ResearchPrincess Alexandra Hospital, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Ken K Y Ho
- Department of EndocrinologyGarvan Institute of Medical Research, St Vincent's Hospital, Sydney, New South Wales 2010, AustraliaSchool of MedicineUniversity of Western Sydney, Sydney, New South Wales 2148, AustraliaThe University of NSWSydney, New South Wales 2052, AustraliaCentres for Health ResearchPrincess Alexandra Hospital, The University of Queensland, Brisbane, Queensland 4102, Australia Department of EndocrinologyGarvan Institute of Medical Research, St Vincent's Hospital, Sydney, New South Wales 2010, AustraliaSchool of MedicineUniversity of Western Sydney, Sydney, New South Wales 2148, AustraliaThe University of NSWSydney, New South Wales 2052, AustraliaCentres for Health ResearchPrincess Alexandra Hospital, The University of Queensland, Brisbane, Queensland 4102, Australia
| |
Collapse
|
13
|
Caglar AS, Kapucu A, Dar KA, Ozkaya HM, Caglar E, Ince H, Kadioglu P. Localization of the aromatase enzyme expression in the human pituitary gland and its effect on growth hormone, prolactin, and thyroid stimulating hormone axis. Endocrine 2015; 49:761-8. [PMID: 25697985 DOI: 10.1007/s12020-015-0537-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/16/2015] [Indexed: 01/27/2023]
Abstract
The aim of this study is to evaluate aromatase expression in prolactin (PRL), thyroid stimulating hormone (TSH), and growth hormone (GH) secreting cells. Nontumoral human pituitary specimens were obtained from autopsy samples. Aromatase co-expression was determined by double immunohistochemical staining and assessed using H scores. H scores for GH-aromatase co-expression (GH-aromatase), TSH-aromatase co-expression (TSH-aromatase), and PRL-aromatase co-expression (PRL-aromatase) were 83.1 ± 13.1, 95.6 ± 16.1, and 83.7 ± 14.5, respectively. TSH producing cells exhibited the highest H score for co-expression of aromatase (p < 0.001). There was no gender difference in terms of H scores for aromatase expression and double immunohistochemical staining results (p > 0.05 for all). There was a negative correlation between the H scores for aromatase and PRL-aromatase, GH-aromatase and TSH-aromatase, respectively (r = -0.592, p < 0.001; r = -0.593, p < 0.001; r = -0.650, p < 0.001, respectively). Also, H scores for aromatase co-expression of each hormone were negatively correlated with the H scores for the corresponding hormone (r = -0.503, p < 0.001 for PRL-aromatase and PRL; r = -0.470, p < 0.001 for GH-aromatase, and GH; r = -0.641, p < 0.001 for TSH-aromatase and TSH). H scores for mean aromatase, GH-aromatase, TSH-aromatase were invariant of age (p > 0.05 for all). Age was negatively correlated with PRL-aromatase H score (r = -0.373, p = 0.008). Our study demonstrated significant aromatase co-expression in PRL, GH, and TSH secreting cells of the human anterior pituitary gland. The mutual paracrinal regulation between aromatase and three adenohypophyseal hormones indicates that aromatase may have a regulatory role on the synthesis and secretion of these hormones.
Collapse
Affiliation(s)
- Asli Sezgin Caglar
- Endocrinology and Metabolism Department, Cerrahpasa Medical School, University of Istanbul, 34303, Cerrahpasa, Istanbul, Turkey
| | | | | | | | | | | | | |
Collapse
|
14
|
Veldhuis JD, Yang RJ, Wigham JR, Erickson D, Miles JC, Bowers CY. Estrogen-like potentiation of ghrelin-stimulated GH secretion by fulvestrant, a putatively selective ER antagonist, in postmenopausal women. J Clin Endocrinol Metab 2014; 99:E2557-64. [PMID: 25210881 PMCID: PMC4255109 DOI: 10.1210/jc.2014-2633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Hyposomatotropism in healthy aging women reflects in part physiological estrogen (estradiol [E2]) depletion associated with menopause. OBJECTIVE AND DESIGN The purpose of this study was to test the hypothesis that low concentrations of endogenous E2 after menopause continue to drive GH secretion. SETTING The study was performed at the Mayo Center for Clinical and Translational Science. PARTICIPANTS The participants were 24 postmenopausal women (aged 50-77 years with body mass index of 19-32 kg/m(2)). INTERVENTIONS This was a randomized, double-blind, placebo-controlled, parallel-cohort treatment study with placebo (PL) (n = 14) or the antiestrogen fulvestrant (FUL) (n = 10) for 3 weeks, followed by infusion of l-arginine with saline, GHRH, ghrelin, or both peptide secretagogues. OUTCOMES GH concentrations were measured over 6 hours with 10-minute sampling and mass spectrometry measures of testosterone, E2, and estrone. RESULTS Concentrations of testosterone, E2, estrone, SHBG, IGF-I, LH, and FSH were not influenced by antiestrogen treatment. In contrast, GH rose from 0.096 ± 0.018 (PL) to 0.23 ± 0.063 μg/L (FUL, P = .033), and IGF-I binding protein type 3 (IGFBP-3) from 3.6 ± 0.18 to 4.0 ± 2.0 mg/L (P = .041). Conversely, prolactin fell from 7.1 ± 0.69 (PL) to 5.5 ± 0.57 μg/L (FUL) (P = .05), and IGF-I binding protein type 1 (IGFBP-1) fell from 44 ± 9.4 to 27 ± 4.3 μg/L (P = .048). Moreover, FUL vs PL potentiated mean GH responses to l-arginine/saline (P = .007), l-arginine/ghrelin (P = .008), and l-arginine/GHRH + ghrelin (P = .031), but not l-arginine/GHRH. CONCLUSION The potent antiestrogen, FUL, amplifies fasting and secretagogue-driven GH secretion and IGFBP-3 concentrations in postmenopausal women without altering SHBG or sex steroid levels. FUL also suppresses prolactin and IGFBP-1, without altering IGF-I. Thus, a major antiestrogen mediates 3 actions of estrogen: agonism (GH), neutral effects (sex steroids), and estrogen antagonism (prolactin and IGFBP-1).
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Endocrine Research Unit (J.D.V., R.J.Y., J.R.W., D.E., J.C.M.), Mayo Clinic College of Medicine Center for Translational Science Activities, Mayo Clinic, Rochester, Minnesota 55905; and Tulane University Health Sciences Center (C.Y.B.), Endocrinology and Metabolism Section, Peptide Research Section, New Orleans, Louisiana 70112
| | | | | | | | | | | |
Collapse
|
15
|
Trifunović S, Manojlović-Stojanoski M, Ajdžanović V, Nestorović N, Ristić N, Medigović I, Milošević V. Effects of genistein on stereological and hormonal characteristics of the pituitary somatotrophs in rats. Endocrine 2014; 47:869-77. [PMID: 24752394 DOI: 10.1007/s12020-014-0265-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/01/2014] [Indexed: 01/08/2023]
Abstract
The hypothalamic-pituitary somatotropic system plays a pivotal role in the regulation of physiological processes and metabolism, which is modulated by gonadal steroids. Considering that genistein belongs to the phytoestrogen family and acts via similar mechanisms to estrogens, the present study was designed to demonstrate whether genistein modulates the morphofunctional characteristic of somatotrophs [growth hormone (GH) cells] in adult rats in comparison with the effects of estradiol. In the study, the orchidectomized adult rats were used as an appropriate model system for testing the effects of this hormone-like substance. Changes in the pituitary somatotrophs were evaluated histologically and stereologically, while GH level was determined biochemically. Using immunolabelling and stereological methods, we showed that orchidectomy (Orx) provoked the decrease of GH cell volume density. After estradiol treatment of Orx rats, the most prominent change concerned the pituitary relative intensity of GH fluorescence and circulating GH level, which were elevated 77 % and 4.7-fold, respectively. Clearly, in contrast to orchidectomy, estradiol treatment enhanced the GH cells activity. Genistein treatment increased pituitary weight and volume, GH cell volume density, the total number of GH cells, and GH blood concentration (1.3-fold) in comparison to the Orx group. Although identical tendencies followed estradiol and genistein administration, the changes observed after genistein treatment were milder compared to estradiol treatment.
Collapse
|
16
|
Abstract
The somatostatin (SRIF) system, which includes the SRIF ligand and receptors, regulates anterior pituitary gland function, mainly inhibiting hormone secretion and to some extent pituitary tumor cell growth. SRIF-14 via its cognate G-protein-coupled receptors (subtypes 1-5) activates multiple cellular signaling pathways including adenylate cyclase/cAMP, MAPK, ion channel-dependent pathways, and others. In addition, recent data have suggested SRIF-independent constitutive SRIF receptor activity responsible for GH and ACTH inhibition in vitro. This review summarizes current knowledge on ligand-dependent and independent SRIF receptor molecular and functional effects on hormone-secreting cells in the anterior pituitary gland.
Collapse
Affiliation(s)
- Tamar Eigler
- Division of EndocrinologyDiabetes and Metabolism, Department of Medicine, Pituitary Center, Cedars Sinai Medical Center, Davis Building, Room 3066, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| | - Anat Ben-Shlomo
- Division of EndocrinologyDiabetes and Metabolism, Department of Medicine, Pituitary Center, Cedars Sinai Medical Center, Davis Building, Room 3066, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| |
Collapse
|
17
|
Norman C, Rollene NL, Erickson D, Miles JM, Bowers CY, Veldhuis JD. Estradiol regulates GH-releasing peptide's interactions with GH-releasing hormone and somatostatin in postmenopausal women. Eur J Endocrinol 2014; 170:121-9. [PMID: 24114435 PMCID: PMC3892701 DOI: 10.1530/eje-13-0733] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Estrogen stimulates pulsatile secretion of GH, via mechanisms that are largely unknown. An untested hypothesis is that estradiol (E₂) drives GH secretion by amplifying interactions among GH-releasing hormone (GHRH), somatostatin (SS), and GH-releasing peptide (GHRP). DESIGN The design comprised double-blind randomized prospective administration of transdermal E₂ vs placebo to healthy postmenopausal women (n=24) followed by pulsatile GHRH or SS infusions for 13 h overnight with or without continuous GHRP2 stimulation. METHODS End points were mean concentrations, deconvolved secretion, and approximate entropy (ApEn; a regularity measure) of GH. RESULTS By generalized ANOVA models, it was observed that E₂ vs placebo supplementation: i) augmented mean (13-h) GH concentrations (P=0.023), GHRH-induced pulsatile GH secretion over the first 3 h (P=0.0085) and pulsatile GH secretion over the next 10 h (P=0.054); ii) increased GHRP-modulated (P=0.022) and SS-modulated (P<0.001) GH ApEn; and iii) did not amplify GHRH/GHRP synergy during pulsatile GH secretion. By linear regression, E₂ concentrations were found to be positively correlated with GH secretion during GHRP2 infusion (P=0.022), whereas BMI was found to be negatively correlated with GH secretion during GHRH (P=0.006) and combined GHRH/GHRP (P=0.015) stimulation. E₂ and BMI jointly determined triple (combined l-arginine, GHRH, and GHRP2) stimulation of GH secretion after saline (R²=0.44 and P=0.003) and pulsatile GHRH (R²=0.39 and P=0.013) infusions. CONCLUSION In summary, in postmenopausal women, E₂ supplementation augments the amount (mass) and alters the pattern (regularity) of GH secretion via interactions among GHRH, SS, GHRP, and BMI. These outcomes introduce a more complex model of E₂ supplementation in coordinating GH secretion in aging women.
Collapse
|
18
|
Norman C, Miles J, Bowers CY, Veldhuis JD. Differential pulsatile secretagogue control of GH secretion in healthy men. Am J Physiol Regul Integr Comp Physiol 2013; 304:R712-9. [PMID: 23485864 DOI: 10.1152/ajpregu.00069.2013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pulsatile growth hormone (GH) secretion putatively reflects integrated regulation by GH-releasing hormone (GHRH), somatostatin (SST), and GH-releasing peptide (GHRP). GHRH and SST secretion is itself pulsatile. However, how GHRH and SST pulses act along with GHRP to jointly determine pulsatile GH secretion is unclear. Moreover, how testosterone (T) modulates such interactions is unknown. These queries were assessed in a prospectively randomized, placebo-controlled double-blind cohort comprising 26 healthy older men randomized to testosterone (T) vs. placebo supplementation. Pulses of GHRH, SST, or saline were infused intravenously at 90-min intervals for 13 h, along with either continuous saline or ghrelin analog (GHRP-2). The train of pulses was followed by a triple stimulus (combined l-arginine, GHRH, and GHRP-2) to estimate near-maximal GH secretion over a final 3 h. Testosterone vs. placebo supplementation doubled pulsatile GH secretion during GHRH pulses combined with continuous saline (GHRH/saline) (P < 0.01). Pulsatile GH secretion correlated positively with T concentrations (270-1,170 ng/dl) in the 26 men during saline pulses/saline (P = 0.015, R(2) = 0.24), GHRH pulses/saline (P = 0.020, R(2) = 0.22), and combined GHRH pulses/GHRP-2 (P = 0.016, R(2) = 0.25) infusions. Basal nonpulsatile GH secretion correlated with T during saline pulses/GHRP-2 drive (P = 0.020, R(2) = 0.16). By regression analysis, pulsatile GH secretion varied negatively with body mass index (BMI) during saline/GHRP-2 infusion (P = 0.001, R(2) = 0.36), as well as after the triple stimulus preceded by GHRH/GHRP-2 (P = 0.013, R(2) = 0.23). Mean (10-h) GH concentrations under GHRP-2 were predicted jointly by estradiol (positively) and BMI (negatively) (P < 0.001, R(2) = 0.520). These data indicate that estradiol, T, and BMI control pulsatile secretagogue-specific GH-regulatory mechanisms in older men.
Collapse
Affiliation(s)
- Catalina Norman
- Endocrine Research Unit, Mayo School of Graduate Medical Education, Center for Translational Science Activities, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
19
|
Darabad RR, Suzuki T, Richards SM, Jensen RV, Jakobiec FA, Zakka FR, Liu S, Sullivan DA. Influence of aromatase absence on the gene expression and histology of the mouse meibomian gland. Invest Ophthalmol Vis Sci 2013; 54:987-98. [PMID: 23233261 DOI: 10.1167/iovs.12-10992] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE We hypothesize that aromatase, an enzyme that controls estrogen biosynthesis, plays a major role in the sex-related differences of the meibomian gland. To begin to test this hypothesis, we examined the influence of aromatase absence, which completely eliminates estrogen production, on glandular gene expression and histology in male and female mice. METHODS Meibomian glands were obtained from adult, age-matched wild-type (WT) and aromatase knockout (ArKO) mice. Tissues were processed for histology or the isolation of total RNA, which was analyzed for differentially expressed mRNAs by using microarrays. RESULTS Our results show that aromatase significantly influences the expression of more than a thousand genes in the meibomian gland. The nature of this effect is primarily sex-dependent. In addition, the influence of aromatase on sex-related differences in gene expression is predominantly genotype-specific. However, many of the sex-related variations in biological process, molecular function, and cellular component ontologies, as well as in KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways, are remarkably similar between WT and ArKO mice. The loss of aromatase activity has no obvious effect on the histology of meibomian glands in male or female mice. CONCLUSIONS Our findings demonstrate that aromatase has a significant impact on gene expression in the meibomian gland. The nature of this influence is sex-dependent and genotype-specific; however, many of the sex-related variations in gene ontologies and KEGG pathways are similar between WT and ArKO mice. Consequently, it appears that aromatase, and by extension estrogen, do not play a major role in the sex-related differences of the mouse meibomian gland.
Collapse
Affiliation(s)
- Raheleh Rahimi Darabad
- Schepens Eye Research Institute, Department of Ophthalmology, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Avtanski D, Novaira HJ, Wu S, Romero CJ, Kineman R, Luque RM, Wondisford F, Radovick S. Both estrogen receptor α and β stimulate pituitary GH gene expression. Mol Endocrinol 2013; 28:40-52. [PMID: 24284820 DOI: 10.1210/me.2013-1245] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although sex steroids have been implicated in the control of mammalian growth, their direct effect on GH synthesis is less clear. The aim of this study was to establish whether estradiol (E2) directly affects GH synthesis in somatotrophs. Somatotroph GH3 and MtT/S cells were used as in vitro models. At physiological doses of E2 stimulation, GH mRNA levels were increased and the ER antagonist ICI 182,780 completely abolished this effect. Estrogen receptor (ER) α- and ERβ-selective agonists, propylpyrazole triol (PPT), and 2,3-bis(4-hydroxyphenyl) propionitrile (DPN), respectively, augmented GH mRNA expression and secretion, whereas E2 and PPT, but not DPN increased prolactin (PRL) mRNA levels. E2, PPT, and DPN stimulated expression of the pituitary transcription factor Pou1f1 and increased its binding to the GH promoter. In vivo evidence of E2 effects on GH synthesis was obtained from the generation of the somatotroph-specific ERα knockout (sERα-KO) mouse model. Basal pituitary GH, PRL, POU1F1, and ERα mRNA expression levels were lower in sERα-KO mice compared with those in controls; whereas ERβ mRNA levels remained unchanged. E2 and DPN stimulated pituitary GH mRNA expression and serum GH levels in control and sERα-KO ovariectomized mice; however, serum GH levels were unchanged in PPT-treated ovariectomized sERα-KO mice. In these animal models, PRL mRNA levels increased after either E2 or PPT, but an increase was not seen after DPN treatment. Thus, we propose a mechanism by which estrogen directly regulates somatotroph GH synthesis at a pretranslational level. In contrast to the predominant effect of ERα in the lactotroph, these results support a role for both ERα and ERβ in the transcriptional control of Gh in the somatotroph and illustrate important differences in ER isoform specificity in the anterior pituitary gland.
Collapse
Affiliation(s)
- Dimiter Avtanski
- Department of Pediatrics (D.A., H.J.N., S.W., C.J.R., S.R.), Division of Endocrinology, and Department of Pediatrics (F.W.), Division of Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Research and Development Division (R.K., R.M.L.), Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Effect of estradiol-17β on follicle-stimulating hormone secretion and egg-laying performance of Japanese quail. Animal 2012; 6:1955-60. [PMID: 23031602 DOI: 10.1017/s1751731112000997] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to measure the effect of estradiol-17β (E2) injection on follicle-stimulating hormone (FSH) secretion and egg-laying performance of Japanese quail. Female Japanese quail were housed in cages and fed ad libitum. After a 7-day adaptation period, the birds were randomly assigned to three groups, that is, one control group and two test groups. The birds were weighed, before every injection. The control group was subcutaneously injected with 0.2 ml sesame oil-ethanol mixture, whereas test groups were injected, twice in a week, with 0.2 ml sesame oil-ethanol mixture containing 0.1 or 0.2 mg E2 along the study. One day after the first injection, egg number, egg weight, eggshell strength and food conception were daily recorded. On the last day of the experiment, the birds were injected and 3 h later seven birds from each group were randomly selected for bleeding. Blood samples (2 ml/bird) were collected from the jugular vein for the measurements of serum concentrations of E2, FSH, calcium (Ca) and phosphorus (P). E2 injection did not cause any significant changes in serum FSH concentrations, daily egg laid/bird, food conception/bird, serum concentrations of the Ca and the P. Egg weight was significantly increased in the 0.1 mg E2-injected group as compared with the control and 0.2 mg E2-injected groups. Eggshell strength in the 0.2 mg E2-injected group was significantly high as compared with the control, whereas the difference between the 0.1 mg E2- and 0.2 mg E2-injected groups was not statistically important. These results show that serum FSH concentration was not increased even when slightly suppressed by subcutaneous injection of 0.1 or 0.2 mg E2. Different doses of E2 have different functions. The increase in BWs in the 0.1 mg E2-injected group was a result of the dose effect, which probably increased growth hormone secretion from the pituitary or IGF-1 synthesis from the liver or both. The dose, 0.2 mg E2, was ineffective in increasing the BW, but it significantly increased eggshell strength probably via the increase in Ca and P utilizations.
Collapse
|
22
|
Schaeffer M, Hodson DJ, Meunier AC, Lafont C, Birkenstock J, Carmignac D, Murray JF, Gavois E, Robinson IC, Le Tissier P, Mollard P. Influence of estrogens on GH-cell network dynamics in females: a live in situ imaging approach. Endocrinology 2011; 152:4789-99. [PMID: 21952249 DOI: 10.1210/en.2011-1430] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The secretion of endocrine hormones from pituitary cells finely regulates a multitude of homeostatic processes. To dynamically adapt to changing physiological status and environmental stimuli, the pituitary gland must undergo marked structural and functional plasticity. Endocrine cell plasticity is thought to primarily rely on variations in cell proliferation and size. However, cell motility, a process commonly observed in a variety of tissues during development, may represent an additional mechanism to promote plasticity within the adult pituitary gland. To investigate this, we used multiphoton time-lapse imaging methods, GH-enhanced green fluorescent protein transgenic mice and sexual dimorphism of the GH axis as a model of divergent tissue demand. Using these methods to acutely (12 h) track cell dynamics, we report that ovariectomy induces a dramatic and dynamic increase in cell motility, which is associated with gross GH-cell network remodeling. These changes can be prevented by estradiol supplementation and are associated with enhanced network connectivity as evidenced by increased coordinated GH-cell activity during multicellular calcium recordings. Furthermore, cell motility appears to be sex-specific, because reciprocal alterations are not detected in males after castration. Therefore, GH-cell motility appears to play an important role in the structural and functional pituitary plasticity, which is evoked in response to changing estradiol concentrations in the female.
Collapse
Affiliation(s)
- Marie Schaeffer
- Centre National de la Recherche Scientifique, Institute of Functional Genomics, Institut National de la Santé et de la Recherche Médicale, Universities of Montpellier 1 and 2, UMR-5203, F-34000 Montpellier, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Birzniece V, Sata A, Sutanto S, Ho KKY. Paracrine regulation of growth hormone secretion by estrogen in women. J Clin Endocrinol Metab 2010; 95:3771-6. [PMID: 20444909 DOI: 10.1210/jc.2010-0476] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Paracrine regulation is emerging as a discrete control mechanism in the endocrine system. In hypogonadal men, stimulation of GH secretion by testosterone requires prior aromatization to estradiol, a paracrine effect unmasked by central estrogen receptor blockade with tamoxifen. In hypogonadal women, estrogen replacement via a physiological non-oral route fails to enhance GH secretion, indicating an absence of an endocrine effect. The aim was to investigate whether local estrogens produced from aromatization regulate GH secretion. DESIGN We conducted an open-label, two-phase, crossover study. PATIENTS AND INTERVENTION We compared the effects on GH secretion of tamoxifen with estradiol valerate in postmenopausal women. Ten women were treated with tamoxifen (10 and 20 mg/d) and estradiol valerate (2 mg/d) via oral route for 2 wk each, with a washout period of at least 6 wk. MAIN OUTCOME MEASURES We measured the GH response to arginine and circulating levels of IGF-I and SHBG, markers of hepatic estrogen effect. RESULTS The GH response to arginine was reduced by 10- and 20-mg tamoxifen in a dose-dependent manner and potentiated significantly (P<0.05) by estradiol valerate. Mean IGF-I concentration was reduced significantly with high-dose tamoxifen (P<0.01) and estradiol valerate treatment (P<0.05), whereas mean SHBG levels rose with both (P<0.01). CONCLUSIONS Blunted GH response to stimulation occurring in the face of reduced IGF-I feedback inhibition with tamoxifen indicates that GH secretion was suppressed by estrogen receptor antagonism. Because circulating estradiol was unaffected, these data indicate a significant role of local estrogen in the central control of GH secretion. We conclude that aromatase mediates the paracrine control of GH secretion in women.
Collapse
Affiliation(s)
- Vita Birzniece
- Garvan Institute of Medical Research, Department of Endocrinology, St. Vincent's Hospital, The University of New South Wales, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
24
|
Rochira V, Zirilli L, Maffei L, Premrou V, Aranda C, Baldi M, Ghigo E, Aimaretti G, Carani C, Lanfranco F. Tall stature without growth hormone: four male patients with aromatase deficiency. J Clin Endocrinol Metab 2010; 95:1626-33. [PMID: 20164294 DOI: 10.1210/jc.2009-1743] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT From preliminary observations, GH-IGF-I seems to be compromised in men with aromatase deficiency. The GH deficiency (GHD) coexists paradoxically with tall stature, raising the question whether or not a true GHD is part of this rare syndrome. OBJECTIVE To evaluate the GH secretion in aromatase-deficient men, their GH response to the GHRH plus arginine (GHRH-ARG) test was compared with that of normal subjects. The effect of estrogen replacement treatment on the GH-IGF-I axis in aromatase-deficient men was evaluated before and during therapy. DESIGN AND SETTING A case-control study was conducted. PATIENTS Four adult men with aromatase deficiency were compared with 12 normal subjects. MAIN OUTCOME MEASURES We measured the GH response to GHRH-ARG in aromatase-deficient men (at baseline and during estrogen treatment) and in normal subjects. Basal serum IGF-I was measured in both patients and controls. RESULTS The response of GH to GHRH-ARG was severely impaired in men with aromatase deficiency and resulted in significantly lower (P < 0.001) levels than in normal subjects. Although normal, serum IGF-I levels were also significantly lower (P < 0.001) than in normal subjects. Both GH peak and IGF-I concentrations were not modified by estrogen therapy in men with aromatase deficiency. CONCLUSIONS In aromatase-deficient men, GH response to potent provocative stimuli is impaired and is not restored by exogenous estrogens. Furthermore, a tall stature may be reached, notwithstanding the coexistence of GHD, if a prolonged time for growth is available due to a delay in bone maturation, and other growth factors different from GH (mainly insulin) promote growth.
Collapse
Affiliation(s)
- Vincenzo Rochira
- Integrated Department of Medicine, Endocrinology, Metabolism, and Geriatrics, Chair of Endocrinology, University of Modena and Reggio Emilia, Via Giardini 1355, 41100 Modena, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Hudson SB, Schroeder DR, Bailey JN, Mielke KL, Erickson D, Miles JM, Bowers CY, Veldhuis JD. Pre- versus postmenopausal age, estradiol, and peptide-secretagogue type determine pulsatile growth hormone secretion in healthy women: studies using submaximal agonist drive and an estrogen clamp. J Clin Endocrinol Metab 2010; 95:353-60. [PMID: 19858315 PMCID: PMC2805499 DOI: 10.1210/jc.2009-1769] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT GH-releasing peptide (GHRP), GHRH, and somatostatin are physiological regulators of pulsatile GH secretion. HYPOTHESIS Age, independently of abdominal visceral fat (AVF) and basal (nonpulsatile) GH secretion, damps pulsatile GH secretion driven by physiological (rather than pharmacological) amounts of GHRP and GHRH in an experimentally controlled estradiol (E(2)) milieu. DESIGN AND SETTING A prospectively randomized, double-blind parallel-cohort study was conducted at an academic medical center. PARTICIPANTS Community-dwelling healthy premenopausal (PRE, age 24 +/- 0.8 yr, n = 20) and postmenopausal (POST, age 63 +/- 1.8 yr, n = 22) women participated in the study. INTERVENTIONS Gonadal-axis down-regulation with leuprolide was followed by randomized addback of placebo or transdermal E(2) and separate-day iv bolus injections of a half-maximally stimulatory dose of GHRP-2 or GHRH (each 0.33 mug/kg). ANALYSIS Three-way analysis of covariance included main factors age, E(2) status, and secretagogue type and covariates AVF and basal GH secretion. RESULTS Submaximally stimulated pulsatile GH secretion was positively determined by PRE vs. POST age (P < 0.001), E(2) repletion vs. depletion (P = 0.001) and GHRP-2 vs. GHRH stimulation (P < 0.001), after adjustment for AVF and basal secretion. E(2) vs. placebo elevated fasting mean GH concentrations in both PRE and POST women (P = 0.006) but increased basal (nonpulsatile) GH secretion in PRE only (P = 0.002). PRE vs. POST age prolonged GHRH-driven GH secretory bursts by 36% (P = 0.006). CONCLUSION PRE vs. POST age, E(2) availability, and physiological peptide drive are triple determinants of pulsatile GH secretion independently of abdominal visceral fat and nonpulsatile GH secretion in healthy women.
Collapse
Affiliation(s)
- Susan B Hudson
- Department of Obstetrics and Gynecology, Mayo School of Graduate Medical Education, Clinical Translational Research Center, Mayo Clinic, Rochester, Minnesota 55905
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Veldhuis JD, Mielke KL, Cosma M, Soares-Welch C, Paulo R, Miles JM, Bowers CY. Aromatase and 5alpha-reductase inhibition during an exogenous testosterone clamp unveils selective sex steroid modulation of somatostatin and growth hormone secretagogue actions in healthy older men. J Clin Endocrinol Metab 2009; 94:973-81. [PMID: 19088159 PMCID: PMC2681279 DOI: 10.1210/jc.2008-2108] [Citation(s) in RCA: 36] [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: 11/19/2022]
Abstract
BACKGROUND How endogenous testosterone (Te), 5alpha-dihydrotestosterone (DHT), and estradiol (E(2)) regulate pulsatile GH secretion is not understood. HYPOTHESIS Conversion of Te to androgenic (Te-->DHT) or estrogenic (Te-->E(2)) products directs GH secretion. SUBJECTS AND LOCATION: Healthy older men (N = 42, ages 50-79 yr) participated at an academic medical center. METHODS We inhibited 5alpha-reduction with dutasteride and aromatization with anastrozole during a pharmacological Te clamp and infused somatostatin (SS), GHRH, GH-releasing peptide-2 (GHRP-2), and L-arginine/GHRH/GHRP-2 (triple stimulus) to modulate GH secretion. ENDPOINTS Deconvolution-estimated basal and pulsatile GH secretion was assessed. RESULTS Administration of Te/placebo elevated Te by 2.8-fold, DHT by 2.6-fold, and E(2) concentrations by 1.9-fold above placebo/placebo. Te/dutasteride and Te/anastrozole reduced stimulated DHT and E(2) by 89 and 86%, respectively. Stepwise forward-selection regression analysis revealed that 1) Te positively determines mean (P = 0.017) and peak (P < 0.001) GH concentrations, basal GH secretion (P = 0.015), and pulsatile GH secretion stimulated by GHRP-2 (P < 0.001); 2) Te and E(2) jointly predict GH responses to the triple stimulus (positively for Te, P = 0.006, and negatively for E(2), P = 0.031); and 3) DHT correlates positively with pulsatile GH secretion during SS infusion (P = 0.011). These effects persisted when abdominal visceral fat was included in the regression. CONCLUSION The present outcomes suggest a tetrapartite model of GH regulation in men, in which systemic concentrations of Te, DHT, and E(2) along with abdominal visceral fat determine the selective actions of GH secretagogues and SS.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Department of Internal Medicine and Pediatrics, Endocrine Research Unit, Clinical Translational Research Center, Mayo Medical and Graduate Schools, Mayo Clinic, Rochester Minnesota 55901, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Veldhuis JD, Keenan DM, Bailey JN, Adeniji A, Miles JM, Paulo R, Cosma M, Soares-Welch C. Testosterone supplementation in older men restrains insulin-like growth factor's dose-dependent feedback inhibition of pulsatile growth hormone secretion. J Clin Endocrinol Metab 2009; 94:246-54. [PMID: 18984660 PMCID: PMC2630862 DOI: 10.1210/jc.2008-1516] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Pulsatile GH secretion declines in older men. The causal mechanisms are unknown. Candidates include deficient feedforward (stimulation) by endogenous secretagogues and excessive feedback (inhibition) by GH or IGF-I due to age and/or relative hypoandrogenemia. HYPOTHESIS Testosterone (T) supplementation in healthy older men will restrain negative feedback by systemic concentrations of IGF-I. SUBJECTS Twenty-four healthy men (ages, 50 to 75 yr; body mass index, 24 to 30 kg/m(2)) participated in the study. METHODS We performed a prospectively randomized, double-blind, placebo-controlled assessment of the impact of pharmacological T supplementation on GH responses to randomly ordered separate-day injections of recombinant human IGF-I doses of 0, 1.0, 1.5, and 2.0 mg/m(2). ANALYSIS Deconvolution and approximate entropy analyses of pulsatile, basal, and entropic (pattern-sensitive) modes of GH secretion were conducted. RESULTS Recombinant human IGF-I injections 1) elevated mean and peak serum IGF-I concentrations dose-dependently (both P < 0.001); 2) suppressed pulsatile GH secretion (P = 0.003), burst mass (P = 0.025), burst number (P = 0.005), interpulse variability (P = 0.032), and basal GH secretion (P = 0.009); and 3) increased secretory pattern regularity (P = 0.020). T administration did not alter experimentally controlled IGF-I concentrations, but it elevated mean GH concentrations (P = 0.015) and stimulated pulsatile GH secretion (frequency P = 0.037, mass per burst P = 0.038). Compared with placebo, T attenuated exogenous IGF-I's inhibition of GH secretory-burst mass (P < 0.038) without restoring pulse number, basal secretion, or pattern regularity. CONCLUSION The capability of systemic T to mute IGF-I feedback on pulsatile GH secretion suggests a novel mechanism for augmenting GH production.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Department of Medicine, Endocrine Research Unit, Mayo School of Graduate Medical Education, Clinical Translational Science Center, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Luque RM, Park S, Kineman RD. Role of endogenous somatostatin in regulating GH output under basal conditions and in response to metabolic extremes. Mol Cell Endocrinol 2008; 286:155-68. [PMID: 18258353 DOI: 10.1016/j.mce.2007.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Revised: 11/28/2007] [Accepted: 12/05/2007] [Indexed: 01/26/2023]
Abstract
Somatostatin (SST) was first described over 30 years ago as a hypothalamic neuropeptide which inhibits GH release. Since that time a large body of literature has accumulated describing how endogenous SST mediates its effects on GH-axis function under normal conditions and in response to metabolic extremes. This review serves to summarize the key findings in this field with a focus on recent progress, much of which has been made possible by the availability of genetically engineered mouse models and SST receptor-specific agonists.
Collapse
Affiliation(s)
- Raul M Luque
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | |
Collapse
|
29
|
Cosma M, Bailey J, Miles JM, Bowers CY, Veldhuis JD. Pituitary and/or peripheral estrogen-receptor alpha regulates follicle-stimulating hormone secretion, whereas central estrogenic pathways direct growth hormone and prolactin secretion in postmenopausal women. J Clin Endocrinol Metab 2008; 93:951-8. [PMID: 18089703 PMCID: PMC2266945 DOI: 10.1210/jc.2007-1322] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Estradiol (E(2)) stimulates GH and prolactin secretion and suppresses FSH secretion in postmenopausal women. Whether central nervous system (CNS) or pituitary mechanisms (or both) mediate such actions is not known. OBJECTIVE Our objective was to distinguish between hypothalamic and pituitary or peripheral (hepatic) actions of E2. SETTING This study was performed in an academic medical center. DESIGN This was a double-blind, prospectively randomized, placebo (Pl)-controlled study. METHODS The capability of a selective, noncompetitive, non-CNS permeant estrogen receptor (ER)-alpha antagonist, fulvestrant (FUL) to antagonize the effects of transdermal E2 and Pl on GH, prolactin, and FSH secretion was assessed in 43 women (ages 50-80 yr) in a four parallel-cohort study. Each woman received four secretagogue infusions to stimulate GH secretion. IGF-I and its binding proteins were measured secondarily. RESULTS Administration of Pl/E2 increased GH and prolactin concentrations by 100%, and suppressed FSH concentrations by more than 50% (each P<or=0.004 compared with Pl/Pl). Treatment with FUL/E2 compared with Pl/E2 partially relieved estrogen's inhibition of FSH secretion (P=0.041), without altering E2's stimulation of prolactin secretion. ANOVA further revealed that: 1) estrogen milieu (P=0.014) and secretagogue type (P<0.001) each determined GH concentrations; 2) FUL/Pl suppressed IGF-I concentrations (P<0.001); 3) FUL abrogated estrogen's elevation of IGF binding protein-1 concentrations (P<0.001); and 4) FUL did not oppose estrogen's suppression of IGF binding protein-3 concentrations (P<0.001). SUMMARY AND CONCLUSIONS Responses to a non-CNS permeant ERalpha antagonist indicate that E2 inhibits FSH secretion in part via pituitary/peripheral ERalpha, drives prolactin output via nonpituitary/nonperipheral-ERalpha effects, and directs GH secretion and IGF-I-binding proteins by complex mechanisms.
Collapse
Affiliation(s)
- Mihaela Cosma
- Endocrine Research Unit, Department of Internal Medicine, Mayo Medical and Graduate Schools of Medicine, Mayo Clinic, 200 First Street S.W., Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
30
|
Frutos MGS, Cacicedo L, Méndez CF, Vicent D, González M, Sánchez-Franco F. Pituitary alterations involved in the decline of growth hormone gene expression in the pituitary of aging rats. J Gerontol A Biol Sci Med Sci 2007; 62:585-97. [PMID: 17595414 DOI: 10.1093/gerona/62.6.585] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Growth hormone (GH) declines during aging. This study investigates whether pituitary constitutive alterations may be involved in the GH decline. Two groups of male Wistar rats were studied (young: 3-month-old; old: 24-month-old). The old rats showed lower pituitary GH messenger RNA (mRNA) levels, immunoreactive rat (IR)-GH content, and GH secretion with no difference in pituitary Pit-1 and cAMP-response element-binding protein (CREB) expression. Pituitary GH releasing hormone receptor (GHRH-R), GH secretagogue receptor (GHS-R), sstr2, and sstr5 mRNA levels were significantly reduced in old rats. The percentage of GH immunoreactive cells was similar in both groups. In vitro, pituitary IR-GH response to GHRH, forskolin (FK), ghrelin, and insulin-like growth factor I (IGF-I) was similar when compared with respective basal secretion and somatostatin-diminished GHRH- and ghrelin-induced IR-GH release in both groups. These results indicate that, as somatotrope function is maintained in aging, the changes observed in GH gene expression and secretion could be reversed by GHS.
Collapse
Affiliation(s)
- Miriam G San Frutos
- Endocrine Service, Hospital Carlos III, C/ Sinesio Delgado, 10, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
31
|
Corman V, Legros JJ. Le traitement hormonal des patients transsexuels et ses conséquences métaboliques. ANNALES D'ENDOCRINOLOGIE 2007; 68:258-64. [PMID: 17693385 DOI: 10.1016/j.ando.2007.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Transsexualism is a sexual identity disorder distinguished by the extreme conviction of belonging to the opposite sex with a total disharmony in the original sex. Diagnosis is established when patients respond to three criteria (DSM-IV): 1) Desire to live and to be accepted as members of opposite sex; 2) Presence of sexual identity disorder for minimal two years; 3) Lack of mental disease or chromosomal anomalies. When diagnosis is confirmed, hormonal treatment can be started and so, improve the secondary sexual characters of selected sex. For patients F-M, treatment is composed of testosterone, most commonly esters of testosterone. For patients M-F, treatment consists of estrogens. These estrogens are frequently associated to an anti-androgen (cyproterone acetate) in the pre-reassignment phase. Avoiding the hepatic way, transdermal form is recommended. Hormonal treatments are not devoid of secondary effects: the most frequent one is venous thromboembolism. Considering contraindications and potential complications, each patient must be selected carefully. The endocrinological follow-up is essential and necessary.
Collapse
Affiliation(s)
- V Corman
- Service d'endocrinologie, CHR Citadelle, 4000 Liège, Belgique
| | | |
Collapse
|
32
|
Veldhuis JD, Keenan DM, Bowers CY. Peripheral estrogen receptor-alpha selectively modulates the waveform of GH secretory bursts in healthy women. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1514-21. [PMID: 17686882 DOI: 10.1152/ajpregu.00438.2007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Estradiol (E(2)) drives growth hormone (GH) secretion via estrogen receptors (ER) located in the hypothalamus and pituitary gland. ERalpha is expressed in GH releasing hormone (GHRH) neurons and GH-secreting cells (somatotropes). Moreover, estrogen regulates receptors for somatostatin, GHR peptide (GHRP, ghrelin), and GH itself, while potentiating signaling by IGF-I. Given this complex network, one cannot a priori predict the selective roles of hypothalamic compared with pituitary ER pathways. To make such a distinction, we introduce an investigative model comprising 1) specific ERalpha blockade with a pure antiestrogen, fulvestrant, that does not penetrate the blood-brain barrier; 2) graded transdermal E(2) administration, which doubles GH concentrations in postmenopausal women; 3) stimulation of fasting GH secretion by pairs of GHRH, GHRP-2 (a ghrelin analog), and l-arginine (to putatively limit somatostatin outflow); and 4) implementation of a flexible waveform deconvolution model to estimate the shape of secretory bursts independently of their size. The combined strategy unveiled that 1) E(2) prolongs GH secretory bursts via fulvestrant-antagonizable mechanisms; 2) fulvestrant extends GHRH/GHRP-2-stimulated secretory bursts; 3) l-arginine/GHRP-2 stimulation lengthens GH secretory bursts whether or not E(2) is present; 4) E(2) limits the capability of l-arginine/GHRP-2 to expand GH secretory bursts, and fulvestrant does not inhibit this effect; and 5) E(2) and/or fulvestrant do not alter the time evolution of l-arginine/GHRH-induced GH secretory bursts. The collective data indicate that peripheral ERalpha-dependent mechanisms determine the shape (waveform) of in vivo GH secretory bursts and that such mechanisms operate with secretagogue selectivity.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Endocrine Research Unit, Mayo Medical and Graduate Schools, Clinical Translational Research Unit, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | |
Collapse
|
33
|
McElvaine AT, Korytko AI, Kilen SM, Cuttler L, Mayo KE. Pituitary-Specific Expression and Pit-1 Regulation of the Rat Growth Hormone-Releasing Hormone Receptor Gene. Mol Endocrinol 2007; 21:1969-83. [PMID: 17536003 DOI: 10.1210/me.2007-0116] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The GHRH receptor is expressed in the somatotroph cell of the anterior pituitary, where it functions to mediate GHRH-stimulated GH release. To study pituitary and somatotroph cell-specific expression of this gene, a transgenic mouse model and complementary cell culture experiments were developed. The activity of the 1.6-kb proximal rat GHRH receptor promoter was examined in vivo by generating transgenic mice with the promoter directing expression of a luciferase reporter. The promoter directs tissue-specific expression; luciferase is highly expressed in the pituitary but absent from 14 other tissues. Immunocytochemistry experiments show that transgene expression is targeted to GH-expressing somatotroph cells. The transgene is 5-fold more highly expressed in males than females, and there is an increase in transgene expression leading up to the onset of puberty. The 1.6-kb promoter was further examined in cell culture experiments, which revealed that the promoter is selectively activated in pituitary cells and that promoter-reporter expression in nonpituitary cells can be enhanced by the pituitary-specific transcription factor Pit-1. EMSAs identified 10 short regions that specifically bind Pit-1 with highly variable relative affinities. The highest affinity site was previously identified and is required for Pit-1 activation of the promoter. Four additional sites contribute to Pit-1 regulation of the promoter and are important to achieving full activation of the gene. The results show that the 1.6-kb promoter is sufficient to direct tissue- and cell-specific expression in vivo and is regulated by Pit-1.
Collapse
Affiliation(s)
- Allison T McElvaine
- Department of Biochemistry, Molecular Biology, and Cell Biology, Northwestern University, Hogan 4-112, Evanston, Illinois 60208, USA
| | | | | | | | | |
Collapse
|
34
|
Veldhuis JD, Cosma M, Erickson D, Paulo R, Mielke K, Farhy LS, Bowers CY. Tripartite control of growth hormone secretion in women during controlled estradiol repletion. J Clin Endocrinol Metab 2007; 92:2336-45. [PMID: 17405836 DOI: 10.1210/jc.2007-0043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Studies of how aging attenuates GH secretion are confounded by differences in sex-steroid milieus, abdominal visceral fat mass (AVF), and IGF-I concentrations and limited in interpretability by the use of pharmacological doses of secretagogues. HYPOTHESIS In a controlled estrogenic milieu, near-physiological secretagogue drive will unmask distinct influences of age, AVF, and IGF-I on GH secretion. LOCATION The study was conducted at an academic medical center. SUBJECTS Subjects included 10 healthy pre- (PRE) and 10 postmenopausal (POST) women. PROCEDURE In a defined estradiol (E(2)) milieu, we compared GH secretion after submaximal stimulation with GH-releasing peptide (GHRP)-2 (ghrelin analog), GHRH, and l-arginine (an inhibitor of somatostatin outflow). ANALYSIS We related GH responses to age stratum (dichotomous variable) and AVF and IGF-I concentrations (continuous variables). RESULTS In the face of comparable concentrations of E(2), testosterone, and SHBG: 1) age (P < 0.001) and secretagogue type (P < 0.001) independently determined GH secretion; 2) GH responses in POST subjects were only 26-33% of those in PRE (P < or = 0.002) across all secretagogues; 3) POST women lost the PRE order of secretagogue potency (GHRP-2 > GHRH = l-arginine); and 4) in the combined cohorts, higher AVF predicted reduced l-arginine-stimulated GH secretion (R(2) = 0.46, P = 0.0013), whereas higher IGF-I concentrations forecast increased GHRP-2 and GHRH drive (R(2) > or = 0.52, P < or = 0.013). CONCLUSION A paradigm of near-physiological secretagogue drive in an E(2)-clamped milieu unmasks tripartite deficits in peptide-signaling pathways in healthy POST, compared with PRE, women. Post hoc analyses indicate that both greater visceral adiposity and lower IGF-I concentrations mark this triple regulatory defect.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Endocrine Research Unit 9, General Clinical Research Center, Mayo Medical and Graduate Schools of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Veldhuis JD, Keenan DM, Bowers CY. Estimation of the size and shape of GH secretory bursts in healthy women using a physiological estradiol clamp and variable-waveform deconvolution model. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1013-21. [PMID: 17537842 DOI: 10.1152/ajpregu.00159.2007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Because estrogen production and age are strong covariates, distinguishing their individual impact on hypothalamo-pituitary regulation of growth hormone (GH) output is difficult. In addition, at fixed elimination kinetics, systemic GH concentration patterns are controlled by three major signal types [GH-releasing hormone (GHRH), GH-releasing peptide (GHRP, ghrelin), and somatostatin (SS)] and by four dynamic mechanisms [the number, mass (size), and shape (waveform) of secretory bursts and basal (time invariant) GH secretion]. The present study introduces an investigative strategy comprising 1) imposition of an experimental estradiol clamp in pre- (PRE) and postmenopausal (POST) women; 2) stimulation of fasting GH secretion by each of GHRH, GHRP-2 (a ghrelin analog), and l-arginine (to putatively limit SSergic restraint); and 3) implementation of a flexible-waveform deconvolution model to estimate basal GH secretion simultaneously with the size and shape of secretory bursts, conditional on pulse number. The combined approach unveiled the following salient percent POST/PRE contrasts: 1) only 27% as much GH secreted in bursts during fasting (P < 0.001); 2) markedly attenuated burstlike GH secretion in response to bolus GHRP-2 (29%), bolus GHRH (30%), l-arginine (37%), constant GHRP-2 (38%), and constant GHRH (42%) (age contrasts, 0.0016 </= P </= 0.027); and 3) a 160% prolongation and 32% abbreviation of the time required to achieve maximal GH secretion after injection of l-arginine and bolus GHRP-2, respectively (both, P < 0.001). Accordingly, age selectively determines both the size (amount) and shape (waveform) of GH secretory bursts in healthy women independently of the short-term estrogen milieu.
Collapse
|
36
|
Vyazovskiy VV, Kopp C, Wigger E, Jones MEE, Simpson ER, Tobler I. Sleep and rest regulation in young and old oestrogen-deficient female mice. J Neuroendocrinol 2006; 18:567-76. [PMID: 16867177 DOI: 10.1111/j.1365-2826.2006.01452.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The effect of circulating oestrogen deficiency on sleep regulation and locomotor activity was investigated in aromatase cytochrome P450 deficient mice (ArKO) and wild-type (WT) controls. Sleep was recorded in 3-month old mice during a 24-h baseline day, 6-h sleep deprivation (SD) and 18-h recovery, and activity was recorded at the age of 3, 9 and 12 months. In mice deficient of oestrogen, the total amount of sleep per 24 h was the same as in WT controls. However, in ArKO mice, sleep was enhanced in the dark period at the expense of sleep in the light phase, and was more fragmented than sleep in WT mice. This redistribution of sleep resulted in a damped amplitude of slow-wave activity (SWA; power between 0.75-4.0 Hz) in non-rapid eye movement sleep across 24 h. After SD, the rebound of sleep and SWA was similar between the genotypes, suggesting that oestrogen deficiency does not affect the mechanisms maintaining the homeostatic balance between the amount of sleep and its intensity. Motor activity decreased with age in both genotypes and was lower in ArKO mice compared to WT at all three ages. After SD, the amount of rest in 3-month old WT mice increased above baseline and was more consolidated. Both effects were less pronounced in ArKO mice, reflecting the baseline differences between the genotypes. The results indicate that despite the pronounced redistribution of sleep and motor activity in oestrogen deficient mice, the basic homeostatic mechanisms of sleep regulation in ArKO mice remain intact.
Collapse
Affiliation(s)
- V V Vyazovskiy
- Institute of Pharmacology and Toxicology, University of Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
37
|
Veldhuis JD, Roemmich JN, Richmond EJ, Bowers CY. Somatotropic and gonadotropic axes linkages in infancy, childhood, and the puberty-adult transition. Endocr Rev 2006; 27:101-40. [PMID: 16434512 DOI: 10.1210/er.2005-0006] [Citation(s) in RCA: 178] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Integrative neuroendocrine control of the gonadotropic and somatotropic axes in childhood, puberty, and young adulthood proceeds via multiple convergent and divergent pathways in the human and experimental animal. Emerging ensemble concepts are required to embody independent, parallel, and interacting mechanisms that subserve physiological adaptations and pathological disruption of reproduction and growth. Significant advances in systems biology will be needed to address these challenges.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Endocrine Research Unit, Department of Internal Medicine, Mayo Medical School, Mayo School of Graduate Medical Education, General Clinical Research Center, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | |
Collapse
|
38
|
Childs GV, Iruthayanathan M, Akhter N, Johnson BW. Estrogen mediated cross talk between the ovary and pituitary somatotrope. Pre-ovulatory support for reproductive activity. Mol Cell Endocrinol 2006; 247:60-3. [PMID: 16443322 PMCID: PMC1751516 DOI: 10.1016/j.mce.2005.12.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Revised: 12/13/2005] [Accepted: 12/14/2005] [Indexed: 11/16/2022]
Affiliation(s)
- Gwen V Childs
- Department of Neurobiology and Developmental Sciences, College of Medicine, 4301 W. Markham, Slot 510, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | | | | | |
Collapse
|
39
|
Iruthayanathan M, Zhou YH, Childs GV. Dehydroepiandrosterone restoration of growth hormone gene expression in aging female rats, in vivo and in vitro: evidence for actions via estrogen receptors. Endocrinology 2005; 146:5176-87. [PMID: 16150906 PMCID: PMC1868401 DOI: 10.1210/en.2005-0811] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A decline in dehydroepiandrosterone (DHEA) and GH levels with aging may be associated with frailty and morbidity. Little is known about the direct effects of DHEA on somatotropes. We recently reported that 17beta-estradiol (E2), a DHEA metabolite, stimulates the expression of GH in vitro in young female rats. To test the hypothesis that DHEA restores function in aging somatotropes, dispersed anterior pituitary (AP) cells from middle-aged (12-14 months) or young (3-4 months) female rats were cultured in vitro with or without DHEA or E2 and fixed for immunolabeling or in situ hybridization. E2 increased the percentage of AP cells with GH protein or mRNA in the aged rats to young levels. DHEA increased the percentages of somatotropes (detected by GH protein or mRNA) from 14-16 +/- 2% to 29-31 +/- 3% (P < or = 0.05) and of GH mRNA (detected by quantitative RT-PCR) only in aging rats. To test DHEA's in vivo effects, 18-month-old female rats were injected with DHEA or vehicle for 2.5 d, followed by a bolus of GHRH 1 h before death. DHEA treatment increased serum GH 1.8-fold (7 +/- 0.5 to 12 +/- 1.3 ng/ml; P = 0.02, by RIA) along with a similar increase (P = 0.02) in GH immunolabel. GHRH target cells also increased from 11 +/- 1% to 19 +/- 2% (P = 0.03). Neither GH nor GHRH receptor mRNAs levels were changed. To test the mechanisms behind DHEA's actions, AP cells from aging rats were treated with DHEA with or without inhibitors of DHEA metabolism. Trilostane, aminogluthemide, or ICI 182,780 completely blocked the stimulatory effects of DHEA, suggesting that DHEA metabolites may stimulate aging somatotropes via estrogen receptors.
Collapse
Key Words
- ap, anterior pituitary
- dhea, dehydroepiandrosterone
- e2, 17β-estradiol
- er, estrogen receptor
- ghrh r, ghrh receptor
- hprt, hypoxanthine guanine phosphoribosyltransferase
- 3β-hsd, 3β-hydroxysteroid dehydrogenase
- iod, integrated optical density
- its, insulin, transferrin, sodium selenite, and bsa
- qrt-pcr, quantitative rt-pcr
Collapse
Affiliation(s)
| | | | - Gwen V. Childs
- Address all correspondence and requests for reprints to: Dr. Gwen V. Childs, Department of Neurobiology and Developmental Sciences, College of Medicine, 4301 W. Markham, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72212. E-mail:
| |
Collapse
|
40
|
Childs GV, Iruthayanathan M, Akhter N, Unabia G, Whitehead-Johnson B. Bipotential effects of estrogen on growth hormone synthesis and storage in vitro. Endocrinology 2005; 146:1780-8. [PMID: 15618363 PMCID: PMC1751515 DOI: 10.1210/en.2004-1111] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Increased pulses of serum GH coincide with rising estrogens during the reproductive cycle, suggesting estrogen regulation. However, there is lack of agreement about estrogen's direct effects on the pituitary. Pituitaries from cycling female rats were dispersed and plated for 24 h in defined media containing vehicle or 0.001-250 nm 17beta-estradiol. Estrogen (0.01-10 nm) increased the percentages of GH antigen-bearing cells in the anterior pituitary significantly (1.3- to 1.6-fold) and 0.01-1 nm concentrations also stimulated significant increases in GH mRNA-bearing cells and in the integrated OD for GH mRNA. However, 100-250 nm either had no effect or, inhibitory effects on the area of label for GH mRNA. To test estrogen's effects on expression of GHRH receptors, cultures were stimulated with biotinylated analogs of GHRH and target cells detected by affinity cytochemistry. Estrogen increased GHRH target cells in populations from rats in all stages of the cycle tested. Basal expression of GHRH target cells declined at metestrus. Cultures treated with 0-1 nm estrogen were then dual labeled for bio-GHRH followed by immunolabeling for GH with the antirabbit IgG-ImmPRESS peroxidase polymer. Over 98% of GH cells bound GHRH and 90-96% of GHRH-bound cells contained GH in all treatment groups. Thus, low concentrations of estrogen may stimulate expression of more cells with GH proteins, biotinylated GHRH binding sites, and GH mRNA, whereas high concentrations have no effect, or may reduce GH mRNA. These bipotential effects may help explain the different findings reported in the literature.
Collapse
Affiliation(s)
- Gwen V Childs
- Department of Neurobiology and Developmental Sciences, College of Medicine, 4301 West Markham, University of Arkansas for Medical Science, Little Rock, Arkansas 72205, USA.
| | | | | | | | | |
Collapse
|
41
|
Kuwahara S, Kesuma Sari D, Tsukamoto Y, Tanaka S, Sasaki F. Age-related changes in growth hormone (GH)-releasing hormone and somatostatin neurons in the hypothalamus and in GH cells in the anterior pituitary of female mice. Brain Res 2005; 1025:113-22. [PMID: 15464751 DOI: 10.1016/j.brainres.2004.08.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2004] [Indexed: 11/16/2022]
Abstract
We have observed growth hormone-releasing hormone (GHRH)-immunoreactive (ir) neurons in the arcuate nucleus (ARC), somatostatin (SS)-ir neurons in the periventricular nucleus (PeN), and pituitary growth hormone (GH)-ir cells in female C57BL/6J mice at 2 months old (2 M), 4, 12 and 23 M, using immunocytochemical and morphometric methods. The number of GHRH-ir neurons decreased with age. The number of SS-ir neurons increased from 2 to 4 M, but decreased after 4 M. The volume of the anterior pituitary and the number of adenohypophysial parenchymal cells fell from 12 to 23 M. The proportion of GH-ir cells decreased significantly from 2 to 4 M and decreased in number from 12 to 23 M as well as in size from 2 to 4 M and from 12 to 23 M. Our results show that both GHRH-ir neurons and SS-ir neurons are fewer in old female mice, but the ratio of the number of SS-ir neurons to GHRH-ir neurons increases in old females. We suggest that the fall in the number and size of GH-ir cells in the pituitary gland with age may be involved in the increase in the ratio of the number of SS-ir neurons to GHRH-ir neurons in the hypothalamus in female mice, as well as in males.
Collapse
Affiliation(s)
- Sachi Kuwahara
- Laboratory of Veterinary Anatomy, Graduate School of Agriculture and Biological Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Sakai, Osaka 599-8531, Japan
| | | | | | | | | |
Collapse
|
42
|
Yan M, Jones MEE, Hernandez M, Liu D, Simpson ER, Chen C. Oestrogen replacement in vivo rescues the dysfunction of pituitary somatotropes in ovariectomised aromatase knockout mice. Neuroendocrinology 2005; 81:158-66. [PMID: 16015026 DOI: 10.1159/000086864] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2004] [Accepted: 04/28/2005] [Indexed: 11/19/2022]
Abstract
The mechanism of regulation of growth hormone (GH) secretion by oestrogens and androgens is still controversial. Available data on the action of oestrogens on GH expression and secretion in somatotropes is poorly understood. We previously reported that the aromatase knockout (ArKO) mouse with oestrogen deficiency and excessive androgen levels had dysfunctional somatotropes. In order to clarify the influence of androgen and oestrogen, we investigated the in vivo treatment of ovariectomised (OVX) ArKO mice with exogenous oestradiol (E2) on the mRNA expression of GH, GH-secretagogue receptor (GHS-R), GH-releasing hormone receptor (GHRH-R), pituitary-specific transcription factor (Pit-1), and somatostatin receptors (sst1-5) in pituitary glands. Circulating plasma GH levels were also evaluated. The results showed that ArKO/OVX mice have a low expression of pituitary GH, GHRH-R, GHS-R and Pit-1, and significantly reduced GH levels. Treatment of female ArKO/OVX (E2-deficient without excessive androgen) mice with E2 for 21 days enhanced expression of pituitary GHRH-R and Pit-1 to 151 and 168%, respectively, of that in mice without treatment. E2 treatment increased GH expression and plasma levels in ArKO/OVX mice to levels comparable with those in wild-type female mice. We conclude therefore that long-term E2 replacement rescues the dysfunction of somatotropes in ArKO/OVX mice through increases in expression of GH, GHRH-R, and Pit-1 in the pituitary somatotropes, whereas the level of androgen in this oestrogen-deficient female mouse does not significantly influence the function of somatotropes.
Collapse
MESH Headings
- Androgens/physiology
- Animals
- Aromatase/genetics
- Aromatase/physiology
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Estradiol/deficiency
- Estradiol/physiology
- Estrogen Replacement Therapy
- Female
- Growth Hormone/genetics
- Growth Hormone/metabolism
- Mice
- Mice, Knockout
- Organ Size
- Ovariectomy
- Pituitary Gland/cytology
- Pituitary Gland/metabolism
- RNA, Messenger/analysis
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Ghrelin
- Receptors, Neuropeptide/genetics
- Receptors, Neuropeptide/metabolism
- Receptors, Pituitary Hormone-Regulating Hormone/genetics
- Receptors, Pituitary Hormone-Regulating Hormone/metabolism
- Receptors, Somatostatin/genetics
- Receptors, Somatostatin/metabolism
- Transcription Factor Pit-1
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Ming Yan
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
43
|
|