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] [Grants] [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
|
Zhao Y, Peng X, Wang Q, Zhang Z, Wang L, Xu Y, Yang H, Bai J, Geng D. Crosstalk Between the Neuroendocrine System and Bone Homeostasis. Endocr Rev 2024; 45:95-124. [PMID: 37459436 DOI: 10.1210/endrev/bnad025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Indexed: 01/05/2024]
Abstract
The homeostasis of bone microenvironment is the foundation of bone health and comprises 2 concerted events: bone formation by osteoblasts and bone resorption by osteoclasts. In the early 21st century, leptin, an adipocytes-derived hormone, was found to affect bone homeostasis through hypothalamic relay and the sympathetic nervous system, involving neurotransmitters like serotonin and norepinephrine. This discovery has provided a new perspective regarding the synergistic effects of endocrine and nervous systems on skeletal homeostasis. Since then, more studies have been conducted, gradually uncovering the complex neuroendocrine regulation underlying bone homeostasis. Intriguingly, bone is also considered as an endocrine organ that can produce regulatory factors that in turn exert effects on neuroendocrine activities. After decades of exploration into bone regulation mechanisms, separate bioactive factors have been extensively investigated, whereas few studies have systematically shown a global view of bone homeostasis regulation. Therefore, we summarized the previously studied regulatory patterns from the nervous system and endocrine system to bone. This review will provide readers with a panoramic view of the intimate relationship between the neuroendocrine system and bone, compensating for the current understanding of the regulation patterns of bone homeostasis, and probably developing new therapeutic strategies for its related disorders.
Collapse
Affiliation(s)
- Yuhu Zhao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaole Peng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Qing Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Zhiyu Zhang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Liangliang Wang
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
- Department of Orthopedics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230022, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| |
Collapse
|
3
|
Laron Z. Challenges in Pediatric Endocrinology. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1757. [PMID: 38002847 PMCID: PMC10670408 DOI: 10.3390/children10111757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023]
Abstract
As the Section Editor-in-Chief, it is my pleasure to introduce the new section of Children devoted to pediatric endocrinology [...].
Collapse
Affiliation(s)
- Zvi Laron
- Endocrine and Diabetes Research Unit, Schneider Children's Medical Center, Tel Aviv University, Petah Tikva 49200, Israel
| |
Collapse
|
4
|
Dong G, Moparthy C, Thome T, Kim K, Yue F, Ryan TE. IGF-1 Therapy Improves Muscle Size and Function in Experimental Peripheral Arterial Disease. JACC Basic Transl Sci 2023; 8:702-719. [PMID: 37426532 PMCID: PMC10322901 DOI: 10.1016/j.jacbts.2022.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 03/11/2023]
Abstract
Lower-extremity peripheral arterial disease (PAD) has increased in prevalence, yet therapeutic development has remained stagnant. Skeletal muscle health and function has been strongly linked to quality of life and medical outcomes in patients with PAD. Using a rodent model of PAD, this study demonstrates that treatment of the ischemic limb with insulin-like growth factor (IGF)-1 significantly increases muscle size and strength without improving limb hemodynamics. Interestingly, the effect size of IGF1 therapy was larger in female mice than in male mice, highlighting the need to carefully examine sex-dependent effects in experimental PAD therapies.
Collapse
Affiliation(s)
- Gengfu Dong
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Chatick Moparthy
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Trace Thome
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Feng Yue
- Department of Animal Sciences, University of Florida, Gainesville, Florida, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
- Center for Exercise Science, University of Florida, Gainesville, Florida, USA
- Myology Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
5
|
Sidhom S, Schneider A, Fang Y, McFadden S, Darcy J, Sathiaseelan R, Palmer AK, Steyn FJ, Grillari J, Kopchick JJ, Bartke A, Siddiqi S, Masternak MM, Stout MB. 17α-Estradiol Modulates IGF1 and Hepatic Gene Expression in a Sex-Specific Manner. J Gerontol A Biol Sci Med Sci 2021; 76:778-785. [PMID: 32857104 PMCID: PMC8087270 DOI: 10.1093/gerona/glaa215] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Indexed: 12/13/2022] Open
Abstract
Aging is the greatest risk factor for most chronic diseases. The somatotropic axis is one of the most conserved biological pathways that regulates aging across species. 17α-Estradiol (17α-E2), a diastereomer of 17β-estradiol (17β-E2), was recently found to elicit health benefits, including improved insulin sensitivity and extend longevity exclusively in male mice. Given that 17β-E2 is known to modulate somatotropic signaling in females through actions in the pituitary and liver, we hypothesized that 17α-E2 may be modulating the somatotropic axis in males, thereby contributing to health benefits. Herein, we demonstrate that 17α-E2 increases hepatic insulin-like growth factor 1 (IGF1) production in male mice without inducing any changes in pulsatile growth hormone (GH) secretion. Using growth hormone receptor knockout (GHRKO) mice, we subsequently determined that the induction of hepatic IGF1 by 17α-E2 is dependent upon GH signaling in male mice, and that 17α-E2 elicits no effects on IGF1 production in female mice. We also determined that 17α-E2 failed to feminize the hepatic transcriptional profile in normal (N) male mice, as evidenced by a clear divergence between the sexes, regardless of treatment. Conversely, significant overlap in transcriptional profiles was observed between sexes in GHRKO mice, and this was unaffected by 17α-E2 treatment. Based on these findings, we propose that 17α-E2 acts as a pleiotropic pathway modulator in male mice by uncoupling IGF1 production from insulin sensitivity. In summary, 17α-E2 treatment upregulates IGF1 production in wild-type (and N) male mice in what appears to be a GH-dependent fashion, while no effects in female IGF1 production are observed following 17α-E2 treatment.
Collapse
Affiliation(s)
- Silvana Sidhom
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rio Grande do Sul, Brazil
| | - Yimin Fang
- Department of Physiology, Southern Illinois University School of Medicine, Springfield
| | - Samuel McFadden
- Department of Physiology, Southern Illinois University School of Medicine, Springfield
| | - Justin Darcy
- Department of Physiology, Southern Illinois University School of Medicine, Springfield
| | - Roshini Sathiaseelan
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center
| | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Frederik J Steyn
- University of Queensland Centre for Clinical Research, Faculty of Medicine, Brisbane, Australia
| | - Johannes Grillari
- Department of Biotechnology, BOKU – University of Natural Resources and Life Sciences, Vienna, Austria
| | - John J Kopchick
- Edison Biotechnology Institute & Heritage College of Osteopathic Medicine, Ohio University, Athens
| | - Andrzej Bartke
- Department of Physiology, Southern Illinois University School of Medicine, Springfield
| | - Shadab Siddiqi
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando
| | - Michael B Stout
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center
- Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center
| |
Collapse
|
6
|
Fang J, Zhang D, Cao JW, Zhang L, Liu CX, Xing YP, Wang F, Xu HY, Wang SC, Ling Y, Wang W, Zhang YR, Zhou HM. Pathways involved in pony body size development. BMC Genomics 2021; 22:58. [PMID: 33461495 PMCID: PMC7814589 DOI: 10.1186/s12864-020-07323-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The mechanism of body growth in mammals is poorly understood. Here, we investigated the regulatory networks involved in body growth through transcriptomic analysis of pituitary and epiphyseal tissues of smaller sized Debao ponies and Mongolian horses at the juvenile and adult stages. RESULTS We found that growth hormone receptor (GHR) was expressed at low levels in long bones, although growth hormone (GH) was highly expressed in Debao ponies compared with Mongolian horses. Moreover, significant downregulated of the GHR pathway components m-RAS and ATF3 was found in juvenile ponies, which slowed the proliferation of bone osteocytes. However, WNT2 and PLCβ2 were obviously upregulated in juvenile Debao ponies, which led to premature mineralization of the bone extracellular matrix. Furthermore, we found that the WNT/Ca2+ pathway may be responsible for regulating body growth. GHR was demonstrated by q-PCR and Western blot analyses to be expressed at low levels in long bones of Debao ponies. Treatment with WNT antagonistI decreased the expression of WNT pathway components (P < 0.05) in vitro. Transduction of ATDC5 cells with a GHR-RNAi lentiviral vector decreased the expression of the GHR pathway components (P < 0.05). Additionally, the expression of the IGF-1 gene in the liver was lower in Debao ponies than in Mongolian horses at the juvenile and adult stages. Detection of plasma hormone concentrations showed that Debao ponies expressed higher levels of IGF-1 as juveniles and higher levels of GH as adults than Mongolian horses, indicating that the hormone regulation in Debao ponies differs from that in Mongolian horses. CONCLUSION Our work provides insights into the genetic regulation of short stature growth in mammals and can provide useful information for the development of therapeutic strategies for small size.
Collapse
Affiliation(s)
- Jun Fang
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Dong Zhang
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Jun Wei Cao
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Li Zhang
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Chun Xia Liu
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Yan Ping Xing
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Feng Wang
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Hong Yang Xu
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Shi Chao Wang
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Yu Ling
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Wei Wang
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China
| | - Yan Ru Zhang
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China.
| | - Huan Min Zhou
- College of Life Sciences, Inner Mongolia Agricultural University, No. 306 Zhaowuda Road, Hohhot, 010018, China.
| |
Collapse
|
7
|
Sarmento-Cabral A, del Rio-Moreno M, Vazquez-Borrego MC, Mahmood M, Gutierrez-Casado E, Pelke N, Guzman G, Subbaiah PV, Cordoba-Chacon J, Yakar S, Kineman RD. GH directly inhibits steatosis and liver injury in a sex-dependent and IGF1-independent manner. J Endocrinol 2021; 248:31-44. [PMID: 33112796 PMCID: PMC7785648 DOI: 10.1530/joe-20-0326] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
A reduction in hepatocyte growth hormone (GH)-signaling promotes non-alcoholic fatty liver disease (NAFLD). However, debate remains as to the relative contribution of the direct effects of GH on hepatocyte function vs indirect effects, via alterations in insulin-like growth factor 1 (IGF1). To isolate the role of hepatocyte GH receptor (GHR) signaling, independent of changes in IGF1, mice with adult-onset, hepatocyte-specific GHR knockdown (aHepGHRkd) were treated with a vector expressing rat IGF1 targeted specifically to hepatocytes. Compared to GHR-intact mice, aHepGHRkd reduced circulating IGF1 and elevated GH. In male aHepGHRkd, the shift in IGF1/GH did not alter plasma glucose or non-esterified fatty acids (NEFA), but was associated with increased insulin, enhanced systemic lipid oxidation and reduced white adipose tissue (WAT) mass. Livers of male aHepGHRkd exhibited steatosis associated with increased de novo lipogenesis, hepatocyte ballooning and inflammation. In female aHepGHRkd, hepatic GHR protein levels were not detectable, but moderate levels of IGF1 were maintained, with minimal alterations in systemic metabolism and no evidence of steatosis. Reconstitution of hepatocyte IGF1 in male aHepGHRkd lowered GH and normalized insulin, whole body lipid utilization and WAT mass. However, IGF1 reconstitution did not reduce steatosis or eliminate liver injury. RNAseq analysis showed IGF1 reconstitution did not impact aHepGHRkd-induced changes in liver gene expression, despite changes in systemic metabolism. These results demonstrate the impact of aHepGHRkd is sexually dimorphic and the steatosis and liver injury observed in male aHepGHRkd mice is autonomous of IGF1, suggesting GH acts directly on the adult hepatocyte to control NAFLD progression.
Collapse
Affiliation(s)
- Andre Sarmento-Cabral
- Department of Medicine, Section of Endocrinology, Diabetes,
and Metabolism, University of Illinois at Chicago and Research and Development
Division, Jesse Brown VA Medical Center, Chicago, IL
| | - Mercedes del Rio-Moreno
- Department of Medicine, Section of Endocrinology, Diabetes,
and Metabolism, University of Illinois at Chicago and Research and Development
Division, Jesse Brown VA Medical Center, Chicago, IL
| | - Mari C. Vazquez-Borrego
- Department of Medicine, Section of Endocrinology, Diabetes,
and Metabolism, University of Illinois at Chicago and Research and Development
Division, Jesse Brown VA Medical Center, Chicago, IL
| | - Mariyah Mahmood
- Department of Medicine, Section of Endocrinology, Diabetes,
and Metabolism, University of Illinois at Chicago and Research and Development
Division, Jesse Brown VA Medical Center, Chicago, IL
| | - Elena Gutierrez-Casado
- Department of Medicine, Section of Endocrinology, Diabetes,
and Metabolism, University of Illinois at Chicago and Research and Development
Division, Jesse Brown VA Medical Center, Chicago, IL
| | - Natalie Pelke
- Department of Medicine, Section of Endocrinology, Diabetes,
and Metabolism, University of Illinois at Chicago and Research and Development
Division, Jesse Brown VA Medical Center, Chicago, IL
| | - Grace Guzman
- Department of Pathology, University of Illinois at Chicago,
College of Medicine, Chicago, IL
| | - Papasani V. Subbaiah
- Department of Medicine, Section of Endocrinology, Diabetes,
and Metabolism, University of Illinois at Chicago and Research and Development
Division, Jesse Brown VA Medical Center, Chicago, IL
| | - Jose Cordoba-Chacon
- Department of Medicine, Section of Endocrinology, Diabetes,
and Metabolism, University of Illinois at Chicago and Research and Development
Division, Jesse Brown VA Medical Center, Chicago, IL
| | - Shoshana Yakar
- Department of Molecular Pathobiology, New York University
College of Dentistry, New York, NY
| | - Rhonda D. Kineman
- Department of Medicine, Section of Endocrinology, Diabetes,
and Metabolism, University of Illinois at Chicago and Research and Development
Division, Jesse Brown VA Medical Center, Chicago, IL
| |
Collapse
|
8
|
Zhang KW, Wu TL, Chen HP, Jiang DN, Zhu CH, Deng SP, Zhang Y, Li GL. Estradiol-17β regulates the expression of insulin-like growth factors 1 and 2 via estradiol receptors in spotted scat (Scatophagus argus). Comp Biochem Physiol B Biochem Mol Biol 2019; 237:110328. [DOI: 10.1016/j.cbpb.2019.110328] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/12/2019] [Accepted: 08/22/2019] [Indexed: 01/17/2023]
|
9
|
Goupille O, Kadri Z, Langelé A, Luccantoni S, Badoual C, Leboulch P, Chrétien S. The integrity of the FOG-2 LXCXE pRb-binding motif is required for small intestine homeostasis. Exp Physiol 2019; 104:1074-1089. [PMID: 31012180 DOI: 10.1113/ep087369] [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: 09/18/2018] [Accepted: 04/16/2019] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Do Fog2Rb- / Rb- mice present a defect of small intestine homeostasis? What is the main finding and its importance? The importance of interactions between FOG-2 and pRb in adipose tissue physiology has previously been demonstrated. Here it is shown that this interaction is also intrinsic to small intestine homeostasis and exerts extrinsic control over mouse metabolism. Thus, this association is involved in maintaining small intestine morphology, and regulating crypt proliferation and lineage differentiation. It therefore affects mouse growth and adaptation to a high-fat diet. ABSTRACT GATA transcription factors and their FOG cofactors play a key role in tissue-specific development and differentiation, from worms to humans. We have shown that GATA-1 and FOG-2 contain an LXCXE pRb-binding motif. Interactions between retinoblastoma protein (pRb) and GATA-1 are crucial for erythroid proliferation and differentiation, whereas the LXCXE pRb-binding site of FOG-2 is involved in adipogenesis. Fog2-knock-in mice have defective pRb binding and are resistant to obesity, due to efficient white-into-brown fat conversion. Our aim was to investigate the pathophysiological impact of FOG-2-pRb interaction on the small intestine and mouse growth. Histological analysis of the small intestine revealed architectural changes in Fog2Rb- / Rb- mice, including villus shortening, with crypt expansion and a change in muscularis propria thickness. These differences were more marked in the proximo-distal part of the small intestine and were associated with an increase in crypt cell proliferation and disruption of the goblet and Paneth cell lineage. The small intestine of the mutants was unable to adapt to a high-fat diet, and had significantly lower plasma lipid levels on such a diet. Fog2Rb- / Rb- mice displayed higher levels of glucose-dependent insulinotropic peptide release, and lower levels of insulin-like growth factor I release on a regular diet. Their intestinal lipid absorption was impaired, resulting in restricted weight gain. In addition to the intrinsic effects of the mutation on adipose tissue, we show here an extrinsic relationship between the intestine and the effect of FOG-2 mutation on mouse metabolism. In conclusion, the interaction of FOG-2 with pRb coordinates the crypt-villus axis and controls small intestine homeostasis.
Collapse
Affiliation(s)
- Olivier Goupille
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Université Paris Sud, Université Paris-Saclay, Fontenay aux Roses, France
| | - Zahra Kadri
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Université Paris Sud, Université Paris-Saclay, Fontenay aux Roses, France
| | - Amandine Langelé
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Université Paris Sud, Université Paris-Saclay, Fontenay aux Roses, France
| | - Sophie Luccantoni
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, Institute of Biology François Jacob, CEA - Université Paris Sud 11 - INSERM U1184, Fontenay-aux-Roses, France
| | - Cécile Badoual
- Department of Pathology, G. Pompidou European Hospital APHP - Université Paris, Descartes, Paris, France
| | - Philippe Leboulch
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Université Paris Sud, Université Paris-Saclay, Fontenay aux Roses, France.,Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Stany Chrétien
- Division of Innovative Therapies, UMR E007, Institute of Biology François Jacob, CEA, Université Paris Sud, Université Paris-Saclay, Fontenay aux Roses, France.,INSERM, Paris, France
| |
Collapse
|
10
|
Kineman RD, del Rio-Moreno M, Sarmento-Cabral A. 40 YEARS of IGF1: Understanding the tissue-specific roles of IGF1/IGF1R in regulating metabolism using the Cre/loxP system. J Mol Endocrinol 2018; 61:T187-T198. [PMID: 29743295 PMCID: PMC7721256 DOI: 10.1530/jme-18-0076] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022]
Abstract
It is clear that insulin-like growth factor-1 (IGF1) is important in supporting growth and regulating metabolism. The IGF1 found in the circulation is primarily produced by the liver hepatocytes, but healthy mature hepatocytes do not express appreciable levels of the IGF1 receptor (IGF1R). Therefore, the metabolic actions of IGF1 are thought to be mediated via extra-hepatocyte actions. Given the structural and functional homology between IGF1/IGF1R and insulin receptor (INSR) signaling, and the fact that IGF1, IGF1R and INSR are expressed in most tissues of the body, it is difficult to separate out the tissue-specific contributions of IGF1/IGF1R in maintaining whole body metabolic function. To circumvent this problem, over the last 20 years, investigators have taken advantage of the Cre/loxP system to manipulate IGF1/IGF1R in a tissue-dependent, and more recently, an age-dependent fashion. These studies have revealed that IGF1/IGF1R can alter extra-hepatocyte function to regulate hormonal inputs to the liver and/or alter tissue-specific carbohydrate and lipid metabolism to alter nutrient flux to liver, where these actions are not mutually exclusive, but serve to integrate the function of all tissues to support the metabolic needs of the organism.
Collapse
Affiliation(s)
- Rhonda D Kineman
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago,1819 W Polk St. M/C 646 Chicago, IL, 60612
- Research and Development Division, Jesse Brown VA Medical Center, Suite 6215, MP 191, 820 S Damen Ave. Chicago, IL 60612
- Corresponding author: Rhonda D Kineman, . University of Illinois at Chicago, Medicine, 1819 W. Polk St., MC 640, Chicago, IL, USA 60612
| | - Mercedes del Rio-Moreno
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago,1819 W Polk St. M/C 646 Chicago, IL, 60612
| | - André Sarmento-Cabral
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago,1819 W Polk St. M/C 646 Chicago, IL, 60612
| |
Collapse
|
11
|
Yakar S, Werner H, Rosen CJ. Insulin-like growth factors: actions on the skeleton. J Mol Endocrinol 2018; 61:T115-T137. [PMID: 29626053 PMCID: PMC5966339 DOI: 10.1530/jme-17-0298] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 12/20/2022]
Abstract
The discovery of the growth hormone (GH)-mediated somatic factors (somatomedins), insulin-like growth factor (IGF)-I and -II, has elicited an enormous interest primarily among endocrinologists who study growth and metabolism. The advancement of molecular endocrinology over the past four decades enables investigators to re-examine and refine the established somatomedin hypothesis. Specifically, gene deletions, transgene overexpression or more recently, cell-specific gene-ablations, have enabled investigators to study the effects of the Igf1 and Igf2 genes in temporal and spatial manners. The GH/IGF axis, acting in an endocrine and autocrine/paracrine fashion, is the major axis controlling skeletal growth. Studies in rodents have clearly shown that IGFs regulate bone length of the appendicular skeleton evidenced by changes in chondrocytes of the proliferative and hypertrophic zones of the growth plate. IGFs affect radial bone growth and regulate cortical and trabecular bone properties via their effects on osteoblast, osteocyte and osteoclast function. Interactions of the IGFs with sex steroid hormones and the parathyroid hormone demonstrate the significance and complexity of the IGF axis in the skeleton. Finally, IGFs have been implicated in skeletal aging. Decreases in serum IGFs during aging have been correlated with reductions in bone mineral density and increased fracture risk. This review highlights many of the most relevant studies in the IGF research landscape, focusing in particular on IGFs effects on the skeleton.
Collapse
Affiliation(s)
- Shoshana Yakar
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010-4086, USA
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Scarborough, Maine 04074, USA
| |
Collapse
|
12
|
Cirmanova V, Zofkova I, Kasalicky P, Lanska V, Bayer M, Starka L, Kanceva R. Hormonal and bone parameters in pubertal girls. Physiol Res 2018; 66:S419-S424. [PMID: 28948826 DOI: 10.33549/physiolres.933733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Here we analyzed associations between muscles mass, total bone mineral content (BMC), lumbar spine bone density (BMD L1-L4) and serum or urine hormones in healthy peripubertal girls. Total BMC and areal BMD L1-L4, muscle mass and fat were measured by dual-energy X-ray absorptiometry (DXA). Muscle force (N) was estimated by a dynamometer. Circulating estradiol, follicle-stimulating hormone (FSH), luteinizing hormone (LH), 25-hydroxy vitamin D, parathyroid hormone (PTH), insulin-like growth factor 1 (IGF-1), leptin, osteocalcin, bone isoenzyme of alkaline phosphatase (bALP) and total calcium and phosphorus were quantified as the nocturnal melatonin and serotonin urinary excretion. Partial correlations adjusted for height, Tanner score and physical activity confirmed positive relationships between BMC or BMD L1-L4 (Z-score) and lean mass or fat. Furthermore, positive relationship was observed between BMC or BMD L1-L4 (Z-score) and serum leptin. After adjustment for Tanner score and physical activity, positive associations were observed between lean mass and IGF-1, leptin levels or muscle force. We proved positive relationships between bone mass and serum leptin in peripubertal girls.
Collapse
Affiliation(s)
- V Cirmanova
- Institute of Endocrinology, Prague, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
13
|
Gilsanz V, Wren TAL, Ponrartana S, Mora S, Rosen CJ. Sexual Dimorphism and the Origins of Human Spinal Health. Endocr Rev 2018; 39:221-239. [PMID: 29385433 PMCID: PMC5888211 DOI: 10.1210/er.2017-00147] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 01/24/2018] [Indexed: 12/26/2022]
Abstract
Recent observations indicate that the cross-sectional area (CSA) of vertebral bodies is on average 10% smaller in healthy newborn girls than in newborn boys, a striking difference that increases during infancy and puberty and is greatest by the time of sexual and skeletal maturity. The smaller CSA of female vertebrae is associated with greater spinal flexibility and could represent the human adaptation to fetal load in bipedal posture. Unfortunately, it also imparts a mechanical disadvantage that increases stress within the vertebrae for all physical activities. This review summarizes the potential endocrine, genetic, and environmental determinants of vertebral cross-sectional growth and current knowledge of the association between the small female vertebrae and greater risk for a broad array of spinal conditions across the lifespan.
Collapse
Affiliation(s)
- Vicente Gilsanz
- Department of Radiology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California 90027.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California 90027.,Department of Orthopaedic Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California 90027
| | - Tishya A L Wren
- Department of Orthopaedic Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California 90027
| | - Skorn Ponrartana
- Department of Radiology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California 90027
| | - Stefano Mora
- Laboratory of Pediatric Endocrinology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Clifford J Rosen
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, Maine 04074
| |
Collapse
|
14
|
Franck SE, Korevaar TIM, Petrossians P, Daly AF, Chanson P, Jaffrain-Réa ML, Brue T, Stalla GK, Carvalho D, Colao A, Hána V, Delemer B, Fajardo C, van der Lely AJ, Beckers A, Neggers SJCMM. A multivariable prediction model for pegvisomant dosing: monotherapy and in combination with long-acting somatostatin analogues. Eur J Endocrinol 2017; 176:421-431. [PMID: 28100630 DOI: 10.1530/eje-16-0956] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/03/2017] [Accepted: 01/17/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Effective treatment of acromegaly with pegvisomant (PEGV), a growth hormone receptor antagonist, requires an appropriate dose titration. PEGV doses vary widely among individual patients, and various covariates may affect its dosing and pharmacokinetics. OBJECTIVE To identify predictors of the PEGV dose required to normalize insulin-like growth factor I (IGF-I) levels during PEGV monotherapy and in combination with long-acting somatostatin analogues (LA-SSAs). DESIGN Two retrospective cohorts (Rotterdam + Liège Acromegaly Survey (LAS), total n = 188) were meta-analyzed as a form of external replication to study the predictors of PEGV dosing in addition to LA-SSA, the LAS (n = 83) was used to study the predictors of PEGV monotherapy dosing. Multivariable regression models were used to identify predictors of the PEGV dose required to normalize IGF-I levels. RESULTS For PEGV dosing in combination with LA-SSA, IGF-I levels, weight, height and age, were associated with the PEGV normalization dosage (P ≤ 0.001, P ≤ 0.001, P = 0.028 and P = 0.047 respectively). Taken together, these characteristics predicted the PEGV normalization dose correctly in 63.3% of all patients within a range of ±60 mg/week (21.3% within a range of ±20 mg/week). For monotherapy, only weight was associated with the PEGV normalization dose (P ≤ 0.001) and predicted this dosage correctly in 77.1% of all patients within a range of ±60 mg/week (31.3% within a range of ±20 mg/week). CONCLUSION In this study, we show that IGF-I levels, weight, height and age can contribute to define the optimal PEGV dose to normalize IGF-I levels in addition to LA-SSA. For PEGV monotherapy, only the patient's weight was associated with the IGF-I normalization PEGV dosage.
Collapse
Affiliation(s)
- S E Franck
- Department of Internal MedicineEndocrinology Section, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - T I M Korevaar
- Department of Internal MedicineEndocrinology Section, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - P Petrossians
- Department of Internal MedicineEndocrinology Section, Centre Hospitalier Universitaire de Liège, University of Liège, Domaine Universitaire du Sart-Tilman, Liège, Belgium
| | - A F Daly
- Department of Internal MedicineEndocrinology Section, Centre Hospitalier Universitaire de Liège, University of Liège, Domaine Universitaire du Sart-Tilman, Liège, Belgium
| | - P Chanson
- Assistance Publique-Hôpitaux de ParisHôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Le Kremlin Bicêtre, France
- Inserm 1185Fac Med Paris Sud, Univ Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - M L Jaffrain-Réa
- Department of Biotechnological and Applied Clinical SciencesUniversity of L'Aquila, L'Aquila and Neuromed, IRCCS, Pozzilli, Italy
| | - T Brue
- Aix-Marseille UniversitéCNRS, CRN2M UMR 7286, Marseille, France
- APHMHôpital Conception, Service d'Endocrinologie, Diabète et Maladies Métaboliques, Centre de Référence des Maladies Rares d'Origine Hypophysaire, Marseille, France
| | - G K Stalla
- Clinical NeuroendocrinologyMax-Planck-Institute of Psychiatry, Munich, Germany
| | - D Carvalho
- Department of EndocrinologyDiabetes and Metabolism Section and Instituto de Investigação e Inovação em Saúde, University of Porto, Centro Hospitalar S. João , Porto, Portugal
| | - A Colao
- Dipartimento di Medicina Clinica e ChirurgiaUniversità Federico II di Napoli, Naples, Italy
| | - V Hána
- 3rd Department of Internal MedicineFirst Medical Faculty, Charles University, Prague, Czech Republic
| | - B Delemer
- Department of EndocrinologyDiabetes, and Nutrition, University Hospital of Reims, Reims, France
| | - C Fajardo
- Servicio de EndocrinologíaHospital Universitario La Ribera, Valencia, Spain
| | - A J van der Lely
- Department of Internal MedicineEndocrinology Section, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A Beckers
- Department of Internal MedicineEndocrinology Section, Centre Hospitalier Universitaire de Liège, University of Liège, Domaine Universitaire du Sart-Tilman, Liège, Belgium
| | - S J C M M Neggers
- Department of Internal MedicineEndocrinology Section, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
15
|
Almeida M, Laurent MR, Dubois V, Claessens F, O'Brien CA, Bouillon R, Vanderschueren D, Manolagas SC. Estrogens and Androgens in Skeletal Physiology and Pathophysiology. Physiol Rev 2017; 97:135-187. [PMID: 27807202 PMCID: PMC5539371 DOI: 10.1152/physrev.00033.2015] [Citation(s) in RCA: 547] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Estrogens and androgens influence the growth and maintenance of the mammalian skeleton and are responsible for its sexual dimorphism. Estrogen deficiency at menopause or loss of both estrogens and androgens in elderly men contribute to the development of osteoporosis, one of the most common and impactful metabolic diseases of old age. In the last 20 years, basic and clinical research advances, genetic insights from humans and rodents, and newer imaging technologies have changed considerably the landscape of our understanding of bone biology as well as the relationship between sex steroids and the physiology and pathophysiology of bone metabolism. Together with the appreciation of the side effects of estrogen-related therapies on breast cancer and cardiovascular diseases, these advances have also drastically altered the treatment of osteoporosis. In this article, we provide a comprehensive review of the molecular and cellular mechanisms of action of estrogens and androgens on bone, their influences on skeletal homeostasis during growth and adulthood, the pathogenetic mechanisms of the adverse effects of their deficiency on the female and male skeleton, as well as the role of natural and synthetic estrogenic or androgenic compounds in the pharmacotherapy of osteoporosis. We highlight latest advances on the crosstalk between hormonal and mechanical signals, the relevance of the antioxidant properties of estrogens and androgens, the difference of their cellular targets in different bone envelopes, the role of estrogen deficiency in male osteoporosis, and the contribution of estrogen or androgen deficiency to the monomorphic effects of aging on skeletal involution.
Collapse
Affiliation(s)
- Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Michaël R Laurent
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Vanessa Dubois
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Frank Claessens
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Charles A O'Brien
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Roger Bouillon
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Dirk Vanderschueren
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas; Departments of Cellular and Molecular Medicine and Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Center for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; and Institut National de la Santé et de la Recherche Médicale UMR1011, University of Lille and Institut Pasteur de Lille, Lille, France
| |
Collapse
|
16
|
Varimo T, Huttunen H, Miettinen PJ, Kariola L, Hietamäki J, Tarkkanen A, Hero M, Raivio T. Precocious Puberty or Premature Thelarche: Analysis of a Large Patient Series in a Single Tertiary Center with Special Emphasis on 6- to 8-Year-Old Girls. Front Endocrinol (Lausanne) 2017; 8:213. [PMID: 28878739 PMCID: PMC5572337 DOI: 10.3389/fendo.2017.00213] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/09/2017] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION We describe the etiology, MRI findings, and growth patterns in girls who had presented with signs of precocious puberty (PP), i.e., premature breast development or early menarche. Special attention was paid to the diagnostic findings in 6- to 8-year-olds. MATERIALS AND METHODS We reviewed the medical records of 149 girls (aged 0.7-10.3 years) who had been evaluated for PP in the Helsinki University Hospital between 2001 and 2014. RESULTS In 6- to 8-year-old girls, PP was most frequently caused by idiopathic gonadotropin-releasing hormone (GnRH)-dependent PP (60%) and premature thelarche (PT; 39%). The former subgroup grew faster (8.7 ± 2.0 cm/year, n = 58) than the girls with PT (7.0 ± 1.1 cm/year, n = 32) (P < 0.001), and the best discrimination for GnRH-dependent PP was achieved with a growth velocity cut-off value of 7.0 cm/year (sensitivity 92% and specificity 58%) [area under the curve 0.82, 95% confidence interval (CI) 0.73-0.91, P < 0.001]. Among asymptomatic and previously healthy 6- to 8-year-old girls with GnRH-dependent PP, one (1.7%, 95% CI 0.3-9.7%) had a pathological brain MRI finding requiring surgical intervention (craniopharyngioma). In girls younger than 3 years, the most frequent cause of breast development was PT, and, in 3- to 6-year-olds, GnRH-dependent PP. CONCLUSION In 6- to 8-year-old girls, analysis of growth velocity is helpful in differentiating between PT and GnRH-dependent PP. Although the frequency of clinically relevant intracranial findings in previously healthy, asymptomatic 6- to 8-year-old girls was low, they can present without any signs or symptoms, which favors routine MRI imaging also in this age group.
Collapse
Affiliation(s)
- Tero Varimo
- Children’s Hospital, Pediatric Research Center, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Heta Huttunen
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Päivi Johanna Miettinen
- Children’s Hospital, Pediatric Research Center, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| | - Laura Kariola
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Johanna Hietamäki
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Annika Tarkkanen
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Matti Hero
- Children’s Hospital, Pediatric Research Center, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Taneli Raivio
- Children’s Hospital, Pediatric Research Center, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki, Finland
- *Correspondence: Taneli Raivio,
| |
Collapse
|
17
|
Yang SO, Wu C, So MY, Lee SJ, Kim YS. Effects of brown rice on cellular growth and metabolic changes in mice. Food Res Int 2016. [DOI: 10.1016/j.foodres.2016.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
18
|
Liu Z, Mohan S, Yakar S. Does the GH/IGF-1 axis contribute to skeletal sexual dimorphism? Evidence from mouse studies. Growth Horm IGF Res 2016; 27:7-17. [PMID: 26843472 PMCID: PMC5488285 DOI: 10.1016/j.ghir.2015.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/24/2015] [Accepted: 12/03/2015] [Indexed: 11/22/2022]
Abstract
The contribution of the gonadotropic axis to skeletal sexual dimorphism (SSD) was clarified in recent years. Studies with animal models of estrogen receptor (ER) or androgen receptor (AR) null mice, as well as mice with bone cell-specific ablation of ER or AR, revealed that both hormones play major roles in skeletal acquisition, and that estrogen regulates skeletal accrual in both sexes. The growth hormone (GH) and its downstream effector, the insulin-like growth factor-1 (IGF-1) are also major determinants of peak bone mass during puberty and young adulthood, and play important roles in maintaining bone integrity during aging. A few studies in both humans and animal models suggest that in addition to the differences in sex steroid actions on bone, sex-specific effects of GH and IGF-1 play essential roles in SSD. However, the contributions of the somatotropic (GH/IGF-1) axis to SSD are controversial and data is difficult to interpret. GH/IGF-1 are pleotropic hormones that act in an endocrine and autocrine/paracrine fashion on multiple tissues, affecting body composition as well as metabolism. Thus, understanding the contribution of the somatotropic axis to SSD requires the use of mouse models that will differentiate between these two modes of action. Elucidation of the relative contribution of GH/IGF-1 axis to SSD is significant because GH is approved for the treatment of normal children with short stature and children with congenital growth disorders. Thus, if the GH/IGF-1 axis determines SSD, treatment with GH may be tailored according to sex. In the following review, we give an overview of the roles of sex steroids in determining SSD and how they may interact with the GH/IGF-1 axis in bone. We summarize several mouse models with impaired somatotropic axis and speculate on the possible contribution of that axis to SSD.
Collapse
Affiliation(s)
- Zhongbo Liu
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology New York University College of Dentistry New York, NY 10010-408, US
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Loma Linda VA Healthcare Systems, Loma Linda, CA 92357
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology New York University College of Dentistry New York, NY 10010-408, US.
| |
Collapse
|
19
|
Juncao C, Pingyang C, Huaxue Q, Danhong H. Puerarin affects bone biomarkers in the serum of rats with intrauterine growth restriction. J TRADIT CHIN MED 2016; 36:211-6. [DOI: 10.1016/s0254-6272(16)30029-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
20
|
Fernández-Pérez L, de Mirecki-Garrido M, Guerra B, Díaz M, Díaz-Chico JC. Sex steroids and growth hormone interactions. ACTA ACUST UNITED AC 2016; 63:171-80. [PMID: 26775014 DOI: 10.1016/j.endonu.2015.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/09/2015] [Accepted: 11/10/2015] [Indexed: 01/17/2023]
Abstract
GH and sex hormones are critical regulators of body growth and composition, somatic development, intermediate metabolism, and sexual dimorphism. Deficiencies in GH- or sex hormone-dependent signaling and the influence of sex hormones on GH biology may have a dramatic impact on liver physiology during somatic development and in adulthood. Effects of sex hormones on the liver may be direct, through hepatic receptors, or indirect by modulating endocrine, metabolic, and gender-differentiated functions of GH. Sex hormones can modulate GH actions by acting centrally, regulating pituitary GH secretion, and peripherally, by modulating GH signaling pathways. The endocrine and/or metabolic consequences of long-term exposure to sex hormone-related compounds and their influence on the GH-liver axis are largely unknown. A better understanding of these interactions in physiological and pathological states will contribute to preserve health and to improve clinical management of patients with growth, developmental, and metabolic disorders.
Collapse
Affiliation(s)
- Leandro Fernández-Pérez
- Institute for Research in Biomedicine and Health (IUIBS), University of Las Palmas de Gran Canaria, Molecular and Translational Pharmacology - BioPharm Group, Las Palmas de G.C., Spain.
| | - Mercedes de Mirecki-Garrido
- Institute for Research in Biomedicine and Health (IUIBS), University of Las Palmas de Gran Canaria, Molecular and Translational Pharmacology - BioPharm Group, Las Palmas de G.C., Spain
| | - Borja Guerra
- Institute for Research in Biomedicine and Health (IUIBS), University of Las Palmas de Gran Canaria, Molecular and Translational Pharmacology - BioPharm Group, Las Palmas de G.C., Spain
| | - Mario Díaz
- Department of Animal Biology, University of La Laguna, Laboratory of Membrane Physiology and Biophysics, La Laguna, Spain
| | - Juan Carlos Díaz-Chico
- Institute for Research in Biomedicine and Health (IUIBS), University of Las Palmas de Gran Canaria, Molecular and Translational Pharmacology - BioPharm Group, Las Palmas de G.C., Spain
| |
Collapse
|
21
|
Garfinkel BP, Arad S, Le PT, Bustin M, Rosen CJ, Gabet Y, Orly J. Proportionate Dwarfism in Mice Lacking Heterochromatin Protein 1 Binding Protein 3 (HP1BP3) Is Associated With Alterations in the Endocrine IGF-1 Pathway. Endocrinology 2015; 156:4558-70. [PMID: 26402843 PMCID: PMC5393342 DOI: 10.1210/en.2015-1668] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/18/2015] [Indexed: 01/08/2023]
Abstract
Heterochromatin protein 1 binding protein 3 (HP1BP3) is a recently described histone H1-related protein with roles in chromatin structure and transcriptional regulation. To explore the potential physiological role of HP1BP3, we have previously described an Hp1bp3(-/-) mouse model with reduced postnatal viability and growth. We now find that these mice are proportionate dwarfs, with reduction in body weight, body length, and organ weight. In addition to their small size, microcomputed tomography analysis showed that Hp1bp3(-/-) mice present a dramatic impairment of their bone development and structure. By 3 weeks of age, mice of both sexes have severely impaired cortical and trabecular bone, and these defects persist into adulthood and beyond. Primary cultures of both osteoblasts and osteoclasts from Hp1bp3(-/-) bone marrow and splenocytes, respectively, showed normal differentiation and function, strongly suggesting that the impaired bone accrual is due to noncell autonomous systemic cues in vivo. One major endocrine pathway regulating both body growth and bone acquisition is the IGF regulatory system, composed of IGF-1, the IGF receptors, and the IGF-binding proteins (IGFBPs). At 3 weeks of age, Hp1bp3(-/-) mice exhibited a 60% reduction in circulating IGF-1 and a 4-fold increase in the levels of IGFBP-1 and IGFBP-2. These alterations were reflected in similar changes in the hepatic transcripts of the Igf1, Igfbp1, and Igfbp2 genes. Collectively, these results suggest that HP1BP3 plays a key role in normal growth and bone development by regulating transcription of endocrine IGF-1 components.
Collapse
Affiliation(s)
- Benjamin P. Garfinkel
- Department of Biological Chemistry (B.P.G., S.A., J.O.), The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; Center for Clinical and Translational Research (P.T.L., C.J.R.), Maine Medical Center Research Institute, Scarborough, Maine 04074; Protein Section (M.B.), Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892; and Department of Anatomy and Anthropology (Y.G.), Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Shiri Arad
- Department of Biological Chemistry (B.P.G., S.A., J.O.), The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; Center for Clinical and Translational Research (P.T.L., C.J.R.), Maine Medical Center Research Institute, Scarborough, Maine 04074; Protein Section (M.B.), Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892; and Department of Anatomy and Anthropology (Y.G.), Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Phuong T. Le
- Department of Biological Chemistry (B.P.G., S.A., J.O.), The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; Center for Clinical and Translational Research (P.T.L., C.J.R.), Maine Medical Center Research Institute, Scarborough, Maine 04074; Protein Section (M.B.), Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892; and Department of Anatomy and Anthropology (Y.G.), Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Michael Bustin
- Department of Biological Chemistry (B.P.G., S.A., J.O.), The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; Center for Clinical and Translational Research (P.T.L., C.J.R.), Maine Medical Center Research Institute, Scarborough, Maine 04074; Protein Section (M.B.), Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892; and Department of Anatomy and Anthropology (Y.G.), Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Clifford J. Rosen
- Department of Biological Chemistry (B.P.G., S.A., J.O.), The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; Center for Clinical and Translational Research (P.T.L., C.J.R.), Maine Medical Center Research Institute, Scarborough, Maine 04074; Protein Section (M.B.), Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892; and Department of Anatomy and Anthropology (Y.G.), Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | | | | |
Collapse
|
22
|
Cordoba-Chacon J, Majumdar N, List EO, Diaz-Ruiz A, Frank SJ, Manzano A, Bartrons R, Puchowicz M, Kopchick JJ, Kineman RD. Growth Hormone Inhibits Hepatic De Novo Lipogenesis in Adult Mice. Diabetes 2015; 64:3093-103. [PMID: 26015548 PMCID: PMC4542445 DOI: 10.2337/db15-0370] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/20/2015] [Indexed: 02/07/2023]
Abstract
Patients with nonalcoholic fatty liver disease (NAFLD) are reported to have low growth hormone (GH) production and/or hepatic GH resistance. GH replacement can resolve the fatty liver condition in diet-induced obese rodents and in GH-deficient patients. However, it remains to be determined whether this inhibitory action of GH is due to direct regulation of hepatic lipid metabolism. Therefore, an adult-onset, hepatocyte-specific, GH receptor (GHR) knockdown (aLivGHRkd) mouse was developed to model hepatic GH resistance in humans that may occur after sexual maturation. Just 7 days after aLivGHRkd, hepatic de novo lipogenesis (DNL) was increased in male and female chow-fed mice, compared with GHR-intact littermate controls. However, hepatosteatosis developed only in male and ovariectomized female aLivGHRkd mice. The increase in DNL observed in aLivGHRkd mice was not associated with hyperactivation of the pathway by which insulin is classically considered to regulate DNL. However, glucokinase mRNA and protein levels as well as fructose-2,6-bisphosphate levels were increased in aLivGHRkd mice, suggesting that enhanced glycolysis drives DNL in the GH-resistant liver. These results demonstrate that hepatic GH actions normally serve to inhibit DNL, where loss of this inhibitory signal may explain, in part, the inappropriate increase in hepatic DNL observed in NAFLD patients.
Collapse
Affiliation(s)
- Jose Cordoba-Chacon
- Research and Development Division, Jesse Brown VA Medical Center, Chicago, IL Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Neena Majumdar
- Research and Development Division, Jesse Brown VA Medical Center, Chicago, IL Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Alberto Diaz-Ruiz
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Stuart J Frank
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL Endocrinology Section Medical Service, Birmingham VA Medical Center, Birmingham, AL
| | - Anna Manzano
- Department of Physiological Sciences, University of Barcelona, L'Hospitalet, Barcelona, Spain
| | - Ramon Bartrons
- Department of Physiological Sciences, University of Barcelona, L'Hospitalet, Barcelona, Spain
| | - Michelle Puchowicz
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Rhonda D Kineman
- Research and Development Division, Jesse Brown VA Medical Center, Chicago, IL Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
23
|
Bennesch MA, Picard D. Minireview: Tipping the balance: ligand-independent activation of steroid receptors. Mol Endocrinol 2015; 29:349-63. [PMID: 25625619 DOI: 10.1210/me.2014-1315] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Steroid receptors are prototypical ligand-dependent transcription factors and a textbook example for allosteric regulation. According to this canonical model, binding of cognate steroid is an absolute requirement for transcriptional activation. Remarkably, the simple one ligand-one receptor model could not be farther from the truth. Steroid receptors, notably the sex steroid receptors, can receive multiple inputs. Activation of steroid receptors by other signals, working through their own signaling pathways, in the absence of the cognate steroids, represents the most extreme form of signaling cross talk. Compared with cognate steroids, ligand-independent activation pathways produce similar but not identical outputs. Here we review the phenomena and discuss what is known about the underlying molecular mechanisms and the biological significance. We hypothesize that steroid receptors may have evolved to be trigger happy. In addition to their cognate steroids, many posttranslational modifications and interactors, modulated by other signals, may be able to tip the balance.
Collapse
Affiliation(s)
- Marcela A Bennesch
- Département de Biologie Cellulaire, Université de Genève, Sciences III, CH-1211 Genève 4, Switzerland
| | | |
Collapse
|
24
|
Connelly KJ, Larson EA, Marks DL, Klein RF. Neonatal estrogen exposure results in biphasic age-dependent effects on the skeletal development of male mice. Endocrinology 2015; 156:193-202. [PMID: 25330099 PMCID: PMC4272395 DOI: 10.1210/en.2014-1324] [Citation(s) in RCA: 14] [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: 01/27/2023]
Abstract
Peak bone mass, one of the most important predictors for fracture risk later in life, is attained during puberty and adolescence and influenced by neonatal and pubertal sex-specific gonadal hormones and GH-IGF-I secretion patterns. This study examined the effects of brief neonatal estrogen (NE) exposure on growth and skeletal development in C57BL/6J mice. A single injection of 100-μg estradiol or vehicle was administered on the first day of life. Growth parameters were monitored and skeletal phenotyping performed at 16 weeks in female mice and at 4 and 16 weeks in male mice. NE exposure negatively impacted adult femoral length in both sexes, but adult body weight, areal bone density, and bone strength in female mice were unaffected. In contrast, somatic growth was attenuated in estrogen-exposed male mice throughout the study period. At the prepubertal time point, the estrogen-exposed males exhibited higher bone mineral density, cortical volume, and cortical thickness compared with controls. However, by the time of peak bone mass acquisition, the early skeletal findings had reversed; estrogen-exposed mice had lower bone density with reduced cross-sectional area, cortical volume, and cortical thickness, resulting in cortical bones that were less resistant to fracture. NE exposure also resulted in reduced testicular volume and lower circulating IGF-I. Male mice exposed to estrogen on the first day of life experience age-dependent changes in skeletal development. Prepubertal animals experience greater endocortical bone acquisition as a result of estrogen exposure. However, by adulthood, continued developmental changes result in overall reduced skeletal integrity.
Collapse
Affiliation(s)
- Kara J Connelly
- Department of Pediatrics (K.J.C., D.L.M.), Division of Pediatric Endocrinology, Oregon Health and Science University Doernbecher Children's Hospital, Portland, Oregon 97239; Bone and Mineral Research Unit (E.A.L., R.F.K.), Department of Medicine, Oregon Health and Science University, Portland, Oregon 97239; Pape Pediatric Research Institute (D.L.M.), Oregon Health and Science University, Portland, Oregon 97239; and Portland Veterans Affairs Medical Center (R.F.K.), Portland, Oregon 97239
| | | | | | | |
Collapse
|
25
|
Vanderschueren D, Laurent MR, Claessens F, Gielen E, Lagerquist MK, Vandenput L, Börjesson AE, Ohlsson C. Sex steroid actions in male bone. Endocr Rev 2014; 35:906-60. [PMID: 25202834 PMCID: PMC4234776 DOI: 10.1210/er.2014-1024] [Citation(s) in RCA: 199] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sex steroids are chief regulators of gender differences in the skeleton, and male gender is one of the strongest protective factors against osteoporotic fractures. This advantage in bone strength relies mainly on greater cortical bone expansion during pubertal peak bone mass acquisition and superior skeletal maintenance during aging. During both these phases, estrogens acting via estrogen receptor-α in osteoblast lineage cells are crucial for male cortical and trabecular bone, as evident from conditional genetic mouse models, epidemiological studies, rare genetic conditions, genome-wide meta-analyses, and recent interventional trials. Genetic mouse models have also demonstrated a direct role for androgens independent of aromatization on trabecular bone via the androgen receptor in osteoblasts and osteocytes, although the target cell for their key effects on periosteal bone formation remains elusive. Low serum estradiol predicts incident fractures, but the highest risk occurs in men with additionally low T and high SHBG. Still, the possible clinical utility of serum sex steroids for fracture prediction is unknown. It is likely that sex steroid actions on male bone metabolism rely also on extraskeletal mechanisms and cross talk with other signaling pathways. We propose that estrogens influence fracture risk in aging men via direct effects on bone, whereas androgens exert an additional antifracture effect mainly via extraskeletal parameters such as muscle mass and propensity to fall. Given the demographic trends of increased longevity and consequent rise of osteoporosis, an increased understanding of how sex steroids influence male bone health remains a high research priority.
Collapse
Affiliation(s)
- Dirk Vanderschueren
- Clinical and Experimental Endocrinology (D.V.) and Gerontology and Geriatrics (M.R.L., E.G.), Department of Clinical and Experimental Medicine; Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine (M.R.L., F.C.); and Centre for Metabolic Bone Diseases (D.V., M.R.L., E.G.), KU Leuven, B-3000 Leuven, Belgium; and Center for Bone and Arthritis Research (M.K.L., L.V., A.E.B., C.O.), Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Chamouni A, Oury F. Reciprocal interaction between bone and gonads. Arch Biochem Biophys 2014; 561:147-53. [DOI: 10.1016/j.abb.2014.06.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/14/2014] [Accepted: 06/17/2014] [Indexed: 11/30/2022]
|
27
|
Tiosano D, Paris F, Grimaldi M, Georgescu V, Servant N, Hochberg Z, Balaguer P, Sultan C. Evidence of ERalpha and ERbeta selectivity and partial estrogen agonism in traditional Chinese medicine. Reprod Biol Endocrinol 2014; 12:97. [PMID: 25300391 PMCID: PMC4201695 DOI: 10.1186/1477-7827-12-97] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 09/07/2014] [Indexed: 11/20/2022] Open
Abstract
The use of complementary and alternative medicine and herbal products, especially traditional Chinese medicines, is progressively rising for both adults and children. This increased use is based on the popular belief that these medicines are safe and harmless. In this report, we describe the results of a bedside-to-bench study that involved a short-statured 4-year-old boy with deficiencies in growth hormone, thyroid stimulating hormone, and adrenocorticotropic hormone due to an ectopic posterior pituitary gland and invisible pituitary stalk. Although the boy was given replacement therapy with hydrocortisone and L-thyroxin, the parents refused to treat him with growth hormone and consulted a naturopath who prescribed a traditional Chinese medicine (TCM) to stimulate the boy's growth. From the age of 20 months, the child's growth was regularly monitored while he was being treated with hydrocortisone, thyroxin, and the TCM. Over a 36-month period, the child's growth velocity accelerated (3 cm/year to 8 cm/year), his height increment substantially increased (-2 SD to -0.8 SD), and his bones matured. In the laboratory investigation, estrogen receptor (ER)alpha and ERbeta reporter cell lines were used to characterize the estrogenic activity of the TCM medicine and its 18 components, and the results established that the medicine and some of its components have estrogen receptor ERalpha and ERbeta selectivity and partial estrogen agonism. Partial estrogenic activity of the TCM was confirmed using whole-cell competitive binding, cell proliferation, and endogenous gene expression assays in the ERalpha-positive breast cancer cell lines. Although the presence of evidence is not always evidence of causality, we have concluded that this traditional Chinese medicine contains ingredients with estrogenic activity that can sustain bone growth and maturation without affecting other estrogen-dependent tissues.
Collapse
Affiliation(s)
- Dov Tiosano
- Pediatric Endocrinology, Meyer Children’s Hospital, Rambam Medical Center, Ruth and Bruce Rappaport Family Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096 Israel
| | - Françoise Paris
- Unité d’Endocrinologie Pédiatrique, CHU Arnaud de Villeneuve, Montpellier, France
- Département d’Hormonologie, CHU Lapeyronie, et Université Montpellier 1, Montpellier, France
- INSERM U896, IRCM, Montpellier, F-34298 France
| | | | - Vera Georgescu
- Département de l’Information Médicale, CHU Montpellier, Montpellier, France
| | - Nadège Servant
- Département d’Hormonologie, CHU Lapeyronie, et Université Montpellier 1, Montpellier, France
| | - Zeev Hochberg
- Pediatric Endocrinology, Meyer Children’s Hospital, Rambam Medical Center, Ruth and Bruce Rappaport Family Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096 Israel
- Rappaport Family Institute for Research in the Medical Sciences, Haifa, 31096 Israel
| | | | - Charles Sultan
- Unité d’Endocrinologie Pédiatrique, CHU Arnaud de Villeneuve, Montpellier, France
- Département d’Hormonologie, CHU Lapeyronie, et Université Montpellier 1, Montpellier, France
| |
Collapse
|
28
|
Abstract
Pubertal maturation plays a fundamental role in bone acquisition. In retrospective epidemiological surveys in pre- and postmenopausal women, relatively later menarcheal age was associated with low bone mineral mass and increased risk of osteoporotic fracture. This association was usually ascribed to shorter time exposure to estrogen from the onset of pubertal maturation to peak bone mass attainment. Recent prospective studies in healthy children and adolescents do not corroborate the limited estrogen exposure hypothesis. In prepubertal girls who will experience later menarche, a reduced bone mineral density was observed before the onset of pubertal maturation, with no further accumulated deficit until peak bone mass attainment. In young adulthood, later menarche is associated with impaired microstructural bone components and reduced mechanical resistance. This intrinsic bone deficit can explain the fact that later menarche increases fracture risk during childhood and adolescence. In healthy individuals, both pubertal timing and bone development share several similar characteristics including wide physiological variability and strong effect of heritable factors but moderate influence of environmental determinants such as nutrition and physical activity. Several conditions modify pubertal timing and bone acquisition, a certain number of them acting in concert on both traits. Taken together, these facts should prompt the search for common genetic regulators of pubertal timing and bone acquisition. It should also open epigenetic investigation avenues to pinpoint which environmental exposure in fetal and infancy life, such as vitamin D, calcium, and/or protein supplies, influences both pubertal timing and bone acquisition.
Collapse
Affiliation(s)
- Jean-Philippe Bonjour
- Division of Bone Diseases, University Hospitals and Faculty of Medicine, CH-1211 Geneva, Switzerland
| | | |
Collapse
|
29
|
Sinnesael M, Claessens F, Laurent M, Dubois V, Boonen S, Deboel L, Vanderschueren D. Androgen receptor (AR) in osteocytes is important for the maintenance of male skeletal integrity: evidence from targeted AR disruption in mouse osteocytes. J Bone Miner Res 2012; 27:2535-43. [PMID: 22836391 DOI: 10.1002/jbmr.1713] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 07/03/2012] [Accepted: 07/09/2012] [Indexed: 11/09/2022]
Abstract
Androgens play a key role in the maintenance of male skeletal integrity. The regulation of this integrity by androgen receptor (AR) signaling has been mainly attributed to osteoblasts. Although osteocytes have emerged as key regulators of bone remodeling, the influence of sex steroids on these cells has been poorly studied. We aimed to investigate the role of AR signaling, specifically in osteocytes using the Cre/LoxP system in male mice (driven by dentin matrix protein 1 [ocy-ARKOs]). Osteocyte fractions of control (AR(ex2)/Y) and ocy-ARKO (ARflox(ex2)/Y; DMP1-cre) mice isolated through sequential collagenase digestion showed increasing AR expression toward the mature osteocyte fraction of control males compared with the more immature fractions, whereas this was reduced by >80% in ocy-ARKO osteocytes. The skeletal phenotype of mutant mice was further assessed by histomorphometry and quantitative micro-computed tomography at 12 and 32 weeks of age. Ocy-ARKOs had significantly lower trabecular bone volume and number in femora and tibias at 32 weeks as well as decreased trabecular number in the L(5) vertebra at 12 weeks. Biomechanical testing showed that ocy-ARKO femora were also stiffer and required a lower ultimate force to induce failure at 32 weeks. However, femoral cortical structure was not significantly different at any time point. The absence of AR in osteocyte also did not appear to affect trabecular bone formation nor its response to mechanical loading. In conclusion, selective inactivation of the AR in osteocytes of male mice accelerates age-related deterioration of skeletal integrity. These findings provide evidence for a direct role of androgens in the maintenance of trabecular bone through actions of the AR in osteocytes.
Collapse
Affiliation(s)
- Mieke Sinnesael
- Clinical and Experimental Endocrinology, Department of Experimental Medicine, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
30
|
The influence of estrogens on the biological and therapeutic actions of growth hormone in the liver. Pharmaceuticals (Basel) 2012; 5:758-78. [PMID: 24281711 PMCID: PMC3763662 DOI: 10.3390/ph5070758] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 07/09/2012] [Accepted: 07/12/2012] [Indexed: 12/13/2022] Open
Abstract
GH is main regulator of body growth and composition, somatic development, intermediate metabolism and gender-dependent dimorphism in mammals. The liver is a direct target of estrogens because it expresses estrogen receptors which are connected with development, lipid metabolism and insulin sensitivity, hepatic carcinogenesis, protection from drug-induced toxicity and fertility. In addition, estrogens can modulate GH actions in liver by acting centrally, regulating pituitary GH secretion, and, peripherally, by modulating GHR-JAK2-STAT5 signalling pathway. Therefore, the interactions of estrogens with GH actions in liver are biologically and clinically relevant because disruption of GH signaling may cause alterations of its endocrine, metabolic, and gender differentiated functions and it could be linked to dramatic impact in liver physiology during development as well as in adulthood. Finally, the interplay of estrogens with GH is relevant because physiological roles these hormones have in human, and the widespread exposition of estrogen or estrogen-related compounds in human. This review highlights the importance of these hormones in liver physiology as well as how estrogens modulate GH actions in liver which will help to improve the clinical use of these hormones.
Collapse
|
31
|
Kerkhofs S, Dubois V, De Gendt K, Helsen C, Clinckemalie L, Spans L, Schuit F, Boonen S, Vanderschueren D, Saunders PTK, Verhoeven G, Claessens F. A role for selective androgen response elements in the development of the epididymis and the androgen control of the 5
α
reductase II gene. FASEB J 2012; 26:4360-72. [DOI: 10.1096/fj.11-202283] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Stefanie Kerkhofs
- Molecular Endocrinology LaboratoryKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| | - Vanessa Dubois
- Molecular Endocrinology LaboratoryKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| | - Karel De Gendt
- Division of Clinical and Experimental EndocrinologyKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| | - Christine Helsen
- Molecular Endocrinology LaboratoryKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| | - Liesbeth Clinckemalie
- Molecular Endocrinology LaboratoryKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| | - Lien Spans
- Molecular Endocrinology LaboratoryKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| | - Frans Schuit
- Gene Expression Unit, Department of Cellular and Molecular MedicineKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| | - Steven Boonen
- Division of Clinical and Experimental EndocrinologyKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| | - Dirk Vanderschueren
- Division of Clinical and Experimental EndocrinologyKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| | - Philippa T. K. Saunders
- Medical Research Council Human Reproductive Sciences UnitThe Queen's Medical Research InstituteEdinburghUK
| | - Guido Verhoeven
- Division of Clinical and Experimental EndocrinologyKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| | - Frank Claessens
- Molecular Endocrinology LaboratoryKatholieke Universiteit LeuvenCampus GasthuisbergLeuvenBelgium
| |
Collapse
|
32
|
Abstract
It has long been known that sex steroid hormones regulate bone mass accrual. This observation raises the testable hypothesis that bone may in turn regulate the synthesis and secretion of sex steroid hormones in one or both genders. This hypothesis is comprised within a more general hypothesis that bone mass, energy metabolism, and reproduction are regulated coordinately. The identification of osteocalcin as an osteoblast-specific secreted molecule allows us to address this question in molecular terms. This review details how the regulation of male fertility by osteocalcin was unraveled, and how osteocalcin signaling in Leydig cells of the testis occurs. It also discusses the implication of this novel mode of regulation of testosterone synthesis observed in males but not in females.
Collapse
Affiliation(s)
- Gerard Karsenty
- Department of Genetics and Development, Columbia University, HHSC 701 West 168th Street, HHSC1602, New York, New York 10032, USA.
| |
Collapse
|
33
|
List EO, Sackmann-Sala L, Berryman DE, Funk K, Kelder B, Gosney ES, Okada S, Ding J, Cruz-Topete D, Kopchick JJ. Endocrine parameters and phenotypes of the growth hormone receptor gene disrupted (GHR-/-) mouse. Endocr Rev 2011; 32:356-86. [PMID: 21123740 PMCID: PMC3365798 DOI: 10.1210/er.2010-0009] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Disruption of the GH receptor (GHR) gene eliminates GH-induced intracellular signaling and, thus, its biological actions. Therefore, the GHR gene disrupted mouse (GHR-/-) has been and is a valuable tool for helping to define various parameters of GH physiology. Since its creation in 1995, this mouse strain has been used by our laboratory and others for numerous studies ranging from growth to aging. Some of the most notable discoveries are their extreme insulin sensitivity in the presence of obesity. Also, the animals have an extended lifespan, which has generated a large number of investigations into the roles of GH and IGF-I in the aging process. This review summarizes the many results derived from the GHR-/- mice. We have attempted to present the findings in the context of current knowledge regarding GH action and, where applicable, to discuss how these mice compare to GH insensitivity syndrome in humans.
Collapse
Affiliation(s)
- Edward O List
- The Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Çolak M, Shimizu T, Matsunaga N, Murayama C, Nagashima S, Kataoka M, Kawashima C, Matsui M, Dorland HAV, Bruckmaier RM, Miyamoto A. Oestradiol Enhances Plasma Growth Hormone and Insulin-like Growth Factor-I Concentrations and Increased the Expression of their Receptors mRNAs in the Liver of Ovariectomized Cows. Reprod Domest Anim 2011; 46:854-61. [DOI: 10.1111/j.1439-0531.2011.01754.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
35
|
Della Torre S, Rando G, Meda C, Stell A, Chambon P, Krust A, Ibarra C, Magni P, Ciana P, Maggi A. Amino acid-dependent activation of liver estrogen receptor alpha integrates metabolic and reproductive functions via IGF-1. Cell Metab 2011; 13:205-14. [PMID: 21284987 DOI: 10.1016/j.cmet.2011.01.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 09/16/2010] [Accepted: 12/30/2010] [Indexed: 11/16/2022]
Abstract
Throughout evolution, organisms have devised strategies to limit fertility in case of prolonged starvation. In mammals, the liver plays a central role in the orchestration of mechanisms allowing for the maintenance of energy homeostasis. We here demonstrate that dietary amino acids regulate the transcriptional activity of hepatic estrogen receptor alpha (ERα) through an mTOR-dependent mechanism. As a result of ERα activation, hepatic IGF-1 mRNA and blood IGF-1 are increased. Conversely, calorie restriction or selective ablation of ERα in the liver decrease blood IGF-1 to levels inadequate for the correct proliferation of the lumen epithelium in the uterus and the progression of the estrous cycle. We propose that the liver acts as critical mediator of energetic and reproductive functions responsible for the blockade of the estrous cycle in case of protein scarcity. Our findings may provide novel insights to understand the cause of selected forms of infertility and metabolic alterations in women after menopause.
Collapse
Affiliation(s)
- Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, 20133, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Olson LE, Ohlsson C, Mohan S. The role of GH/IGF-I-mediated mechanisms in sex differences in cortical bone size in mice. Calcif Tissue Int 2011; 88:1-8. [PMID: 21113585 PMCID: PMC3042806 DOI: 10.1007/s00223-010-9436-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 10/31/2010] [Indexed: 12/20/2022]
Abstract
Cortical bone dimensions are important determinants of bone strength. Gender differences in cortical bone size caused by greater periosteal expansion in males than in females during the pubertal growth spurt are well established both in humans and in experimental animal models. However, the mechanism by which gender influences cortical bone size is still a matter of investigation. The role of androgens and estrogen in pubertal bone growth has been examined in human disorders as well as animal models, such as gonadectomized or sex steroid receptor knockout mice. Based on the findings that growth hormone (GH) and insulin-like growth factor I (IGF-I) are major regulators of postnatal skeletal growth, we and others have predicted that sex hormones interact with the GH/IGF-I axis to regulate cortical bone size. However, studies conflict as to whether estrogen and androgens impact cortical bone size through the canonical pathway, through GH without IGF-I mediation, through IGF-I without GH stimulation, or independent of GH/IGF-I. We review recent data on the impact of sex steroids and components of the GH/IGF axis on sexual dimorphism in bone size. While the GH/IGF-I axis is a major player in regulating peak bone size, the relative contribution of GH/IGF-dependent mechanisms to sex differences in cortical bone size remains to be established.
Collapse
Affiliation(s)
- Lisa E Olson
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center and Loma Linda University, Loma Linda, CA 92357, USA
| | | | | |
Collapse
|
37
|
Van Vaerenbergh I, Blockeel C, Van Lommel L, Ghislain V, In't Veld P, Schuit F, Fatemi HM, Devroey P, Bourgain C. Cyclooxygenase-2 network as predictive molecular marker for clinical pregnancy in in vitro fertilization. Fertil Steril 2011; 95:448-51, 451.e1-2. [DOI: 10.1016/j.fertnstert.2010.07.1061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 07/01/2010] [Accepted: 07/14/2010] [Indexed: 11/28/2022]
|
38
|
Bauer-Dantoin AC, Meinhardt DJ. 17β-Estradiol Exposure Accelerates Skeletal Development in Xenopus laevis Tadpoles. Anat Rec (Hoboken) 2010; 293:1880-6. [DOI: 10.1002/ar.21226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Angela C Bauer-Dantoin
- Human Biology Program and Department of Biology, University of Wisconsin - Green Bay, Green Bay, Wisconsin 54311-7001, USA.
| | | |
Collapse
|
39
|
Sexual dimorphism in cortical bone size and strength but not density is determined by independent and time-specific actions of sex steroids and IGF-1: evidence from pubertal mouse models. J Bone Miner Res 2010; 25:617-26. [PMID: 19888832 DOI: 10.1359/jbmr.090828] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although it is well established that males acquire more bone mass than females, the underlying mechanism and timing of this sex difference remain controversial. The aim of this study was to assess the relative contribution of sex steroid versus growth hormone-insulin-like growth factor 1 (GH-IGF-1) action to pubertal bone mass acquisition longitudinally in pubertal mice. Radial bone expansion peaked during early puberty (3 to 5 weeks of age) in male and female mice, with significantly more expansion in males than in females (+40%). Concomitantly, in 5 week old male versus female mice, periosteal and endocortical bone formation was higher (+70%) and lower (-47%), respectively, along with higher serum IGF-1 levels during early puberty in male mice. In female mice, ovariectomy increased radial bone expansion during early puberty as well as the endocortical perimeter. In male mice, orchidectomy reduced radial bone expansion only during late puberty (5 to 8 weeks of age), whereas combined androgen and estrogen deficiency modestly decreased radial bone expansion during early puberty, accompanied by lower IGF-1 levels. GHRKO mice with very low IGF-1 levels, on the other hand, showed limited radial bone expansion and no skeletal dimorphism. From these data we conclude that skeletal sexual dimorphism is established during early puberty and depends primarily on GH-IGF-1 action. In males, androgens and estrogens have stimulatory effects on bone size during late and early puberty, respectively. In females, estrogens limit bone size during early puberty. These longitudinal findings in mice provide strong evidence that skeletal dimorphism is determined by independent and time-specific effects of sex steroids and IGF-1.
Collapse
|
40
|
Vico L, Vanacker JM. Sex hormones and their receptors in bone homeostasis: insights from genetically modified mouse models. Osteoporos Int 2010; 21:365-72. [PMID: 19495826 DOI: 10.1007/s00198-009-0963-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 04/29/2009] [Indexed: 10/20/2022]
Abstract
In this review, we summarize available data regarding bone phenotypes in estrogen receptors alpha and beta, androgen receptor, and aromatase enzyme-deficient mice. We examine sex differences in the trabecular and cortical bone compartments and we discuss these findings in relation to known estrogen effects in humans. We also report how estrogen influences the responsiveness of the skeleton to exercise. Although uncertainties remain, it is clear that both estrogen and androgen are important for both male and female skeleton. Estrogen receptor alpha mainly through its classical signaling pathway is particularly important for the male mice skeleton while both estrogen receptors alpha and beta are required for female mice skeleton. These deletions also induce major hormonal alterations themselves impacting on bone metabolism. More investigations are needed to fully understand the respective role of all these receptors in periosteal expansion in both sexes and the way they affect the mechanical sensitivity of the periosteum.
Collapse
Affiliation(s)
- L Vico
- Université de Lyon, INSERM, U890, Université Jean Monnet, St-Etienne, 42023, France.
| | | |
Collapse
|
41
|
Callewaert F, Boonen S, Vanderschueren D. Sex steroids and the male skeleton: a tale of two hormones. Trends Endocrinol Metab 2010; 21:89-95. [PMID: 19837603 DOI: 10.1016/j.tem.2009.09.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2009] [Revised: 09/04/2009] [Accepted: 09/09/2009] [Indexed: 10/20/2022]
Abstract
Traditionally, the stronger male skeleton was considered to result from higher androgen levels in men compared to women. However, the regulation of male bone growth by sex steroids appears more complex than originally anticipated. Based on clinical observations and studies in animal models, not only androgens and androgen receptor (AR), but also estrogens and estrogen receptor-alpha (not ERbeta) are required for optimal bone mineral acquisition during male growth. In addition, both sex steroids are involved in the maintenance of male skeletal health. In fact, bone loss and fracture risk have been associated with estrogen exposure in elderly men. Overall, a compelling body of evidence suggests that both androgens and estrogens are crucial for male skeletal growth and maintenance.
Collapse
Affiliation(s)
- Filip Callewaert
- Department of Experimental Medicine, Katholieke Universiteit Leuven, Center for Musculoskeletal Research, Leuven University, Herestraat 49, B-3000 Belgium
| | | | | |
Collapse
|
42
|
Amuzie CJ, Pestka JJ. Suppression of insulin-like growth factor acid-labile subunit expression--a novel mechanism for deoxynivalenol-induced growth retardation. Toxicol Sci 2009; 113:412-21. [PMID: 19805407 DOI: 10.1093/toxsci/kfp225] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Consumption of deoxynivalenol (DON), a trichothecene mycotoxin commonly detected in cereal-based foods, causes impaired growth in many animal species. While growth retardation is used as a basis for regulating DON levels in human food, the underlying mechanisms remain poorly understood. Oral exposure of mice to DON rapidly induces multiorgan expression of proinflammatory cytokines, and this is followed by upregulation of several suppressors of cytokine signaling (SOCS), some of which are capable of impairing growth hormone (GH) signaling. The purpose of this study was to test the hypothesis that impairment of the GH axis precedes DON-induced growth retardation in the mouse. Subchronic dietary exposure of young (4-week old) mice to DON (20 ppm) over a period of 2-8 weeks was found to (1) impair weight gain, (2) result in a steady-state plasma DON concentration (40-60 ng/ml), (3) downregulate hepatic insulin-like growth factor acid-labile subunit (IGFALS) mRNA expression, and (4) reduce circulating insulin-like growth factor 1 (IGF1) and IGFALS levels. Acute oral exposure to DON at 0.5-12.5 mg/kg body weight (bw) markedly suppressed hepatic IGFALS mRNA levels within 2 h in a dose-dependent fashion, whereas 0.1 mg/kg bw was without effect. DON-induced IGFALS mRNA upregulation occurred both with and without exogenous GH treatment. These latter effects co-occurred with robust hepatic suppressors of cytokine signaling 3 upregulation. Taken together, these data suggest that oral DON exposure perturbs GH axis by suppressing two clinically relevant growth-related proteins, IGFALS and IGF1. Both have potential to serve as biomarkers of effect in populations exposed to this common foodborne mycotoxin.
Collapse
Affiliation(s)
- Chidozie J Amuzie
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, Michigan 48824, USA
| | | |
Collapse
|
43
|
Windahl SH, Andersson N, Chagin AS, Mårtensson UEA, Carlsten H, Olde B, Swanson C, Movérare-Skrtic S, Sävendahl L, Lagerquist MK, Leeb-Lundberg LMF, Ohlsson C. The role of the G protein-coupled receptor GPR30 in the effects of estrogen in ovariectomized mice. Am J Physiol Endocrinol Metab 2009; 296:E490-6. [PMID: 19088255 DOI: 10.1152/ajpendo.90691.2008] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In vitro studies suggest that the membrane G protein-coupled receptor GPR30 is a functional estrogen receptor (ER). The aim of the present study was to determine the possible in vivo role of GPR30 as a functional ER primarily for the regulation of skeletal parameters, including bone mass and longitudinal bone growth, but also for some other well-known estrogen-regulated parameters, including uterine weight, thymus weight, and fat mass. Three-month-old ovariectomized (OVX) GPR30-deficient mice (GPR30(-/-)) and wild-type (WT) mice were treated with either vehicle or increasing doses of estradiol (E(2); 0, 30, 70, 160, or 830 ng.mouse(-1).day(-1)). Body composition [bone mineral density (BMD), fat mass, and lean mass] was analyzed by dual-energy-X ray absorptiometry, while the cortical and trabecular bone compartments were analyzed by peripheral quantitative computerized tomography. Quantitative histological analyses were performed in the distal femur growth plate. Bone marrow cellularity and distribution were analyzed using a fluorescence-activated cell sorter. The estrogenic responses on most of the investigated parameters, including increase in bone mass (total body BMD, spine BMD, trabecular BMD, and cortical bone thickness), increase in uterine weight, thymic atrophy, fat mass reduction, and increase in bone marrow cellularity, were similar for all of the investigated E(2) doses in WT and GPR30(-/-) mice. On the other hand, E(2) treatment reduced longitudinal bone growth, reflected by decreased femur length and distal femur growth plate height, in the WT mice but not in the GPR30(-/-) mice compared with vehicle-treated mice. These in vivo findings demonstrate that GPR30 is not required for normal estrogenic responses on several major well-known estrogen-regulated parameters. In contrast, GPR30 is required for a normal estrogenic response in the growth plate.
Collapse
Affiliation(s)
- S H Windahl
- Institute of Medicine, Sahlgrenska Academy, Göteborg University, Göteborg
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Van Vaerenbergh I, Van Lommel L, Ghislain V, In't Veld P, Schuit F, Fatemi HM, Devroey P, Bourgain C. In GnRH antagonist/rec-FSH stimulated cycles, advanced endometrial maturation on the day of oocyte retrieval correlates with altered gene expression. Hum Reprod 2009; 24:1085-91. [DOI: 10.1093/humrep/den501] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
45
|
Li L, He D, Wilborn TW, Falany JL, Falany CN. Increased SULT1E1 activity in HepG2 hepatocytes decreases growth hormone stimulation of STAT5b phosphorylation. Steroids 2009; 74:20-9. [PMID: 18831980 PMCID: PMC2633718 DOI: 10.1016/j.steroids.2008.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Revised: 08/21/2008] [Accepted: 09/02/2008] [Indexed: 11/20/2022]
Abstract
Mouse models of cystic fibrosis (CF) display increased sulfotransferase 1E1 (SULT1E1) activity in hepatocytes of cystic fibrosis transmembrane receptor (CFTR)-deficient animals. SULT1E1 is responsible for the sulfation and inactivation of beta-estradiol (E2) at physiological concentrations. IGF-1 message levels in CFTR(-/-) mouse livers were positively correlated with body weight and negatively correlated with SULT1E1 activity. Growth hormone (GH) is important in the regulation of hepatic IGF-1 expression indicating that E2 levels are involved with GH signaling in hepatocytes. To investigate the effects of E2 and SULT1E1 activity on GH signal transduction in human hepatocytes, SULT1E1 was stably expressed in HepG2 cells. Effects of increased E2 sulfation on the GH signaling pathway and E2-regulated gene expression were examined. Pretreatment of HepG2 cells with 10nM E2 prior to GH stimulation increased STAT5b phosphorylation and IGF-1 expression. In SULT1E1-transfected HepG2 cells, GH-stimulated STAT5b phosphorylation was significantly decreased. E2 treatment had no effect on STAT5b phosphorylation in the absence of GH stimulation. E2 also had no effect on Jak-2 phosphorylation. E2 has an apparent rapid action on increasing GH-stimulated STAT5b phosphorylation that was not attenuated by the estrogen receptor antagonist, ICI 182,780. Physiological levels of E2 in HepG2 cells increase GH stimulation of IGF-1 production apparently through increased phosphorylated STAT5b levels and transcriptional activation of the IGF-1 gene. The enhanced SULT1E1 activity may have a role in inhibiting GH-stimulated STAT5b phosphorylation and IGF-1 synthesis via the sulfation and inactivation of E2.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Dongning He
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Teresa W. Wilborn
- Department of Pharmaceutical Sciences, Samford University, Birmingham, AL 35229, United States
| | - Josie L. Falany
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Charles N. Falany
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| |
Collapse
|
46
|
Govoni KE, Wergedal JE, Chadwick RB, Srivastava AK, Mohan S. Prepubertal OVX increases IGF-I expression and bone accretion in C57BL/6J mice. Am J Physiol Endocrinol Metab 2008; 295:E1172-80. [PMID: 18812464 PMCID: PMC2584807 DOI: 10.1152/ajpendo.90507.2008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Accepted: 09/21/2008] [Indexed: 11/22/2022]
Abstract
It is generally well accepted that the pubertal surge in estrogen is responsible for the rapid bone accretion that occurs during puberty and that this effect is mediated by an estrogen-induced increase in growth hormone (GH)/insulin-like growth factor (IGF) action. To test the cause and effect relationship between estrogen and GH/IGF, we evaluated the consequence of ovariectomy (OVX) in prepubertal mice (C57BL/6J mice at 3 wk of age) on skeletal changes and the GH/IGF axis during puberty. Contrary to our expectations, OVX increased body weight (12-18%), bone mineral content (11%), bone length (4%), bone size (3%), and serum, liver, and bone IGF-I (30-50%) and decreased total body fat (18%) at 3 wk postsurgery. To determine whether estrogen is the key ovarian factor responsible for these changes, we performed a second experiment in which OVX mice were treated with placebo or estrogen implants. In addition to observing similar results compared with our first experiment, estrogen treatment partially rescued the increased body weight and bone size and completely rescued body fat and IGF-I levels. The increased bone accretion in OVX mice was due to increased bone formation rate (as determined by bone histomorphometry) and increased serum procollagen peptide. In conclusion, contrary to the known estrogen effect as an initiator of GH/IGF surge and thereby pubertal growth spurt, our findings demonstrate that loss of estrogen and/or other hormones during the prepubertal growth period effect leads to an increase in IGF-I production and bone accretion in mice.
Collapse
Affiliation(s)
- Kristen E Govoni
- Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California 92357, USA
| | | | | | | | | |
Collapse
|
47
|
Kamanga-Sollo E, White ME, Chung KY, Johnson BJ, Dayton WR. Potential role of G-protein-coupled receptor 30 (GPR30) in estradiol-17beta-stimulated IGF-I mRNA expression in bovine satellite cell cultures. Domest Anim Endocrinol 2008; 35:254-62. [PMID: 18650055 DOI: 10.1016/j.domaniend.2008.06.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 05/14/2008] [Accepted: 06/02/2008] [Indexed: 10/21/2022]
Abstract
Androgenic and estrogenic steroids enhance muscle growth in animals and humans. Estradiol-17beta (E2) and trenbolone acetate (TBA) (a synthetic testosterone analog) increased IGF-I mRNA expression in bovine muscle satellite cell (BSC) cultures. The goal of this study was to evaluate the mechanisms responsible for this increase by evaluating the effects of ICI 182 780 (an E2 receptor antagonist), flutamide (an androgen receptor inhibitor), G1 (a GPR30 agonist), and BSA-conjugated E2 on E2 and/or TBA-stimulated IGF-I mRNA expression in BSC cultures. Flutamide completely suppressed TBA-stimulated IGF-I mRNA expression in BSC cultures. ICI 182 780 did not suppress E2-stimulated IGF-I mRNA expression and 100 nM ICI 182 780 enhanced (93%, p<0.05) IGF-I mRNA levels in BSC cultures. G1 (100 nM) stimulated IGF-I mRNA expression (100%, p<0.05) but had no effect on proliferation in BSC cultures. E2-BSA, which cannot cross the cell membrane, stimulated IGF-I mRNA expression (approximately 100%, p<0.05) in BSC but even at extremely high concentrations had no effect on proliferation. In summary, our data indicate the E2-stimulation of proliferation and E2-stimulation of IGF-I mRNA expression in BSC cultures occur via different mechanisms. Our previous results showing that ICI 182 780 inhibited BSC proliferation and results of the current study showing lack of response to E2-BSA or G1 suggest that E2-stimulated proliferation in BSC cultures is mediated through classical estrogen receptors. Stimulation by ICI 182 780, G1 and E2-BSA suggests the E2-stimulated IGF-I mRNA expression in BSC cultures is mediated through the GPR30 receptor.
Collapse
MESH Headings
- Androgen Antagonists/pharmacology
- Animals
- Cattle/physiology
- Cell Proliferation/drug effects
- Cyclin G
- Cyclin G1
- Cyclins/pharmacology
- Estradiol/analogs & derivatives
- Estradiol/pharmacology
- Estrogen Antagonists/pharmacology
- Flutamide/pharmacology
- Fulvestrant
- Insulin-Like Growth Factor I/biosynthesis
- Insulin-Like Growth Factor I/genetics
- Male
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, G-Protein-Coupled/metabolism
- Reverse Transcriptase Polymerase Chain Reaction/veterinary
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/drug effects
- Satellite Cells, Skeletal Muscle/metabolism
- Serum Albumin, Bovine/pharmacology
- Trenbolone Acetate/analogs & derivatives
- Trenbolone Acetate/pharmacology
Collapse
Affiliation(s)
- E Kamanga-Sollo
- Animal Growth and Development Laboratory, Department of Animal Science, University of Minnesota, 348 ABLMS, Eckles Avenue, St. Paul, MN 55108, USA
| | | | | | | | | |
Collapse
|
48
|
Callewaert F, Venken K, Ophoff J, De Gendt K, Torcasio A, van Lenthe GH, Van Oosterwyck H, Boonen S, Bouillon R, Verhoeven G, Vanderschueren D. Differential regulation of bone and body composition in male mice with combined inactivation of androgen and estrogen receptor-alpha. FASEB J 2008; 23:232-40. [PMID: 18809737 DOI: 10.1096/fj.08-113456] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Osteoporosis and muscle frailty are important health problems in elderly men and may be partly related to biological androgen activity. This androgen action can be mediated directly through stimulation of the androgen receptor (AR) or indirectly through stimulation of estrogen receptor-alpha (ERalpha) following aromatization of androgens into estrogens. To assess the differential action of AR and ERalpha pathways on bone and body composition, AR-ERalpha double-knockout mice were generated and characterized. AR disruption decreased trabecular bone mass, whereas ERalpha disruption had no additional effect on the AR-dependent trabecular bone loss. In contrast, combined AR and ERalpha inactivation additionally reduced cortical bone and muscle mass compared with either AR or ERalpha disruption alone. ERalpha inactivation--in the presence or absence of AR--increased fat mass. We demonstrate that AR activation is solely responsible for the development and maintenance of male trabecular bone mass. Both AR and ERalpha activation, however, are needed to optimize the acquisition of cortical bone and muscle mass. ERalpha activation alone is sufficient for the regulation of fat mass. Our findings clearly define the relative importance of AR and ERalpha signaling on trabecular and cortical bone mass as well as body composition in male mice.
Collapse
Affiliation(s)
- Filip Callewaert
- Laboratory for Experimental Medicine and Endocrinology, Department of Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Srivastava VK, Hiney JK, Mattison JA, Bartke A, Dees WL. The Alcohol-Induced Suppression of Ovarian Insulin-Like Growth Factor-1 Gene Transcription Is Independent of Growth Hormone and Its Receptor. Alcohol Clin Exp Res 2007; 31:880-6. [PMID: 17386070 DOI: 10.1111/j.1530-0277.2007.00368.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Insulin-like growth factor-1 (IGF-1) plays an important role in ovarian development and function. Alcohol (ALC) is a gonadal toxin and capable of causing depressed ovarian IGF-1 and suppressed estradiol. The mechanism by which ALC affects IGF-1 transcription is not well understood, and more information is needed to better understand the interrelationships between ALC, growth hormone (GH) and its ovarian receptor, and the gene expression of ovarian IGF-1. METHODS Prepubertal transgenic mice carrying the bovine GH (bGH) gene were fed either a liquid diet containing ALC, pair-fed the companion isocaloric control liquid diet, or fed chow and water. A fourth group consisted of normal (nontransgenic) littermates fed chow and water. Mice received their diets for 5 days, were then killed and tissues collected and frozen. RESULTS Alcohol did not alter circulating levels of bGH held constant by the promoter. Real-time polymerase chain reaction (PCR) showed elevated (p<0.05) ovarian IGF-1 mRNA levels in both groups of transgenic control mice, compared with normal mice. Insulin-like growth factor-1 expression in the ALC-treated transgenic mice was suppressed (p<0.01) compared with both transgenic controls. Insulin-like growth factor-1 receptor (IGF-1R) gene expression was also decreased (p<0.01) in ALC-treated transgenic mice compared with transgenic controls. Growth hormone-receptor (GH-R) synthesis revealed that all transgenic mice, including those exposed to ALC, showed increased (p<0.05) GH-R mRNA compared with normal controls, and ALC did not alter protein levels of the GH-R. CONCLUSIONS These results suggest that the ALC-induced suppression of ovarian IGF-1 gene transcription is independent of alterations in serum GH.
Collapse
Affiliation(s)
- Vinod K Srivastava
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A & M University, College Station, Texas 77843-4458, USA.
| | | | | | | | | |
Collapse
|
50
|
Bibliography. Current world literature. Growth and development. Curr Opin Endocrinol Diabetes Obes 2007; 14:74-89. [PMID: 17940424 DOI: 10.1097/med.0b013e32802e6d87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|