1
|
Ryu K, Lee O, Roh J. Age-related changes in densitometry and histomorphometry of long bone during the pubertal growth spurt in male rats. Anat Sci Int 2024; 99:268-277. [PMID: 38598056 DOI: 10.1007/s12565-024-00766-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/09/2024] [Indexed: 04/11/2024]
Abstract
Because experimental studies to determine the developmental toxicity of exposure to various substances in children are impossible, many studies use immature male rats. This study aimed to provide normative data for longitudinal bone growth with age during the puberty in male rats. In order to evaluate long bone growth and mineralization we examined bone size and bone density by dual-energy X-ray absorptiometry, analyzed histomorphometry of the growth plate, and serum hormone levels relevant to bone growth from postnatal day (PD)20 to PD60. The length and weight of long bones increased strongly by PD40, and no further increase was observed after PD50. On the other hand, tibial growth plate height decreased sharply after PD50 along with a reduction in the number of cells and columns, which was probably responsible for the absence of further lengthening of long bones. Parameters related to bone formation such as bone area ratio, and the thickness and number of trabeculae, also increased significantly between PD40 and PD50. Furthermore, serum levels of IGF-1 peaked at PD30 and testosterone increased rapidly on and after PD40, when IGF-1 levels were going down. These changes may participate in the parallel increase in mineral acquisition, as well as lengthening of long bones. Our findings provide comprehensive data for changes in bone density, histomorphometry of long bones, and hormone levels relevant to bone growth during the growth spurt. This will be useful for planning animal toxicological studies, particularly for deciding on the appropriate age of animals to use in given experiments.
Collapse
Affiliation(s)
- Kiyoung Ryu
- Department of Obstetrics & Gynecology, College of Medicine, Hanyang University, Guri, 11923, South Korea
| | - Okto Lee
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul, 133-791, South Korea
| | - Jaesook Roh
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul, 133-791, South Korea.
| |
Collapse
|
2
|
Kuraku S, Kaiya H, Tanaka T, Hyodo S. Evolution of Vertebrate Hormones and Their Receptors: Insights from Non-Osteichthyan Genomes. Annu Rev Anim Biosci 2023; 11:163-182. [PMID: 36400012 DOI: 10.1146/annurev-animal-050922-071351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Homeostatic control and reproductive functions of humans are regulated at the molecular levels largely by peptide hormones secreted from endocrine and/or neuroendocrine cells in the central nervous system and peripheral organs. Homologs of those hormones and their receptors function similarly in many vertebrate species distantly related to humans, but the evolutionary history of the endocrine system involving those factors has been obscured by the scarcity of genome DNA sequence information of some taxa that potentially contain their orthologs. Focusing on non-osteichthyan vertebrates, namely jawless and cartilaginous fishes, this article illustrates how investigating genome sequence information assists our understanding of the diversification of vertebrate gene repertoires in four broad themes: (a) the presence or absence of genes, (b) multiplication and maintenance of paralogs, (c) differential fates of duplicated paralogs, and (d) the evolutionary timing of gene origins.
Collapse
Affiliation(s)
- Shigehiro Kuraku
- Molecular Life History Laboratory, Department of Genomics and Evolutionary Biology, National Institute of Genetics, Mishima, Japan; .,Department of Genetics, Sokendai (Graduate University for Advanced Studies), Mishima, Japan.,Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan
| | - Hiroyuki Kaiya
- Grandsoul Research Institute of Immunology, Inc., Uda, Japan
| | - Tomohiro Tanaka
- Department of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Susumu Hyodo
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Japan
| |
Collapse
|
3
|
Evidences for Mutant Huntingtin Inducing Musculoskeletal and Brain Growth Impairments via Disturbing Testosterone Biosynthesis in Male Huntington Disease Animals. Cells 2022; 11:cells11233779. [PMID: 36497038 PMCID: PMC9737670 DOI: 10.3390/cells11233779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/11/2022] [Accepted: 11/20/2022] [Indexed: 11/29/2022] Open
Abstract
Body weight (BW) loss and reduced body mass index (BMI) are the most common peripheral alterations in Huntington disease (HD) and have been found in HD mutation carriers and HD animal models before the manifestation of neurological symptoms. This suggests that, at least in the early disease stage, these changes could be due to abnormal tissue growth rather than tissue atrophy. Moreover, BW and BMI are reported to be more affected in males than females in HD animal models and patients. Here, we confirmed sex-dependent growth alterations in the BACHD rat model for HD and investigated the associated contributing factors. Our results showed growth abnormalities along with decreased plasma testosterone and insulin-like growth factor 1 (IGF-1) levels only in males. Moreover, we demonstrated correlations between growth parameters, IGF-1, and testosterone. Our analyses further revealed an aberrant transcription of testosterone biosynthesis-related genes in the testes of BACHD rats with undisturbed luteinizing hormone (LH)/cAMP/PKA signaling, which plays a key role in regulating the transcription process of some of these genes. In line with the findings in BACHD rats, analyses in the R6/2 mouse model of HD showed similar results. Our findings support the view that mutant huntingtin may induce abnormal growth in males via the dysregulation of gene transcription in the testis, which in turn can affect testosterone biosynthesis.
Collapse
|
4
|
Young JA, Zhu S, List EO, Duran-Ortiz S, Slama Y, Berryman DE. Musculoskeletal Effects of Altered GH Action. Front Physiol 2022; 13:867921. [PMID: 35665221 PMCID: PMC9160929 DOI: 10.3389/fphys.2022.867921] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/25/2022] [Indexed: 12/17/2022] Open
Abstract
Growth hormone (GH) is a peptide hormone that can signal directly through its receptor or indirectly through insulin-like growth factor 1 (IGF-1) stimulation. GH draws its name from its anabolic effects on muscle and bone but also has distinct metabolic effects in multiple tissues. In addition to its metabolic and musculoskeletal effects, GH is closely associated with aging, with levels declining as individuals age but GH action negatively correlating with lifespan. GH’s effects have been studied in human conditions of GH alteration, such as acromegaly and Laron syndrome, and GH therapies have been suggested to combat aging-related musculoskeletal diseases, in part, because of the decline in GH levels with advanced age. While clinical data are inconclusive, animal models have been indispensable in understanding the underlying molecular mechanisms of GH action. This review will provide a brief overview of the musculoskeletal effects of GH, focusing on clinical and animal models.
Collapse
Affiliation(s)
- Jonathan A. Young
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Shouan Zhu
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
- Ohio Musculoskeletal and Neurological Institute, Heritage College of Osteopathic Medicine, Athens, OH, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Edward O. List
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | | | - Yosri Slama
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Darlene E. Berryman
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
- Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- *Correspondence: Darlene E. Berryman,
| |
Collapse
|
5
|
Sarcopenia in Children with Solid Organ Tumors: An Instrumental Era. Cells 2022; 11:cells11081278. [PMID: 35455957 PMCID: PMC9024674 DOI: 10.3390/cells11081278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/22/2022] [Accepted: 04/05/2022] [Indexed: 11/17/2022] Open
Abstract
Sarcopenia has recently been studied in both adults and children and was found to be a prognostic marker for adverse outcome in a variety of patient groups. Our research showed that sarcopenia is a relevant marker in predicting outcome in children with solid organ tumors, such as hepatoblastoma and neuroblastoma. This was especially true in very ill, high-risk groups. Children with cancer have a higher likelihood of ongoing loss of skeletal muscle mass due to a mismatch in energy intake and expenditure. Additionally, the effects of cancer therapy, hormonal alterations, chronic inflammation, multi-organ dysfunction, and a hypermetabolic state all contribute to a loss of skeletal muscle mass. Sarcopenia seems to be able to pinpoint this waste to a high degree in a new and objective way, making it an additional tool in predicting and improving outcome in children. This article focuses on the current state of sarcopenia in children with solid organ tumors. It details the pathophysiological mechanisms behind sarcopenia, highlighting the technical features of the available methods for measuring muscle mass, strength, and function, including artificial intelligence (AI)-based techniques. It also reviews the latest research on sarcopenia in children, focusing on children with solid organ tumors.
Collapse
|
6
|
Ong C, Lee JH, Leow MKS, Puthucheary ZA. A narrative review of skeletal muscle atrophy in critically ill children: pathogenesis and chronic sequelae. Transl Pediatr 2021; 10:2763-2777. [PMID: 34765499 PMCID: PMC8578782 DOI: 10.21037/tp-20-298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/18/2020] [Indexed: 11/10/2022] Open
Abstract
Muscle wasting is now recognized as a growing, debilitating problem in critically ill adults, resulting in long-term deficits in function and an impaired quality of life. Ultrasonography has demonstrated decreases in skeletal muscle size during pediatric critical illness, although variations exist. However, muscle protein turnover patterns during pediatric critical illness are unclear. Understanding muscle protein turnover during critical illness is important in guiding interventions to reduce muscle wasting. The aim of this review was to explore the possible protein synthesis and breakdown patterns in pediatric critical illness. Muscle protein turnover studies in critically ill children are lacking, with the exception of those with burn injuries. Children with burn injuries demonstrate an elevation in both muscle protein breakdown (MPB) and synthesis during critical illness. Extrapolations from animal models and whole-body protein turnover studies in children suggest that children may be more dependent on anabolic factors (e.g., nutrition and growth factors), and may experience greater muscle degradation in response to insults than adults. Yet, children, particularly the younger ones, are more responsive to anabolic agents, suggesting modifiable muscle wasting during critical illness. There is a lack of evidence for muscle wasting in critically ill children and its correlation with outcomes, possibly due to current available methods to study muscle protein turnover in children-most of which are invasive or tedious. In summary, children may experience muscle wasting during critical illness, which may be more reversible by the appropriate anabolic agents than adults. Age appears an important determinant of skeletal muscle turnover. Less invasive methods to study muscle protein turnover and associations with long-term outcome would strengthen the evidence for muscle wasting in critically ill children.
Collapse
Affiliation(s)
- Chengsi Ong
- Nutrition and Dietetics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Jan Hau Lee
- Children's Intensive Care Unit, KK Women's Children's Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Melvin K S Leow
- Duke-NUS Medical School, Singapore, Singapore.,Clinical Nutrition Research Center, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Zudin A Puthucheary
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| |
Collapse
|
7
|
miR-19-3p Promotes Autophagy and Apoptosis in Pelvic Organ Prolapse Through the AKT/mTOR/p70S6K Pathway: Function of miR-19-3p on Vaginal Fibroblasts by Targeting IGF-1. Female Pelvic Med Reconstr Surg 2021; 27:e630-e638. [PMID: 34432732 PMCID: PMC8389349 DOI: 10.1097/spv.0000000000001034] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Pelvic organ prolapse (POP) is a common condition in older women. A decrease in collagen 1 (Col-1) expression is one of the main causes of POP. Many microRNAs play an important role in regulating target genes. The relationship between miR-19-3p and POP is investigated in this study, and the molecular mechanism was also explored to find whether miR-19-3p may be a potential target for early diagnosis and prevention of POP. METHODS A total of 60 patients with POP and 60 patients without POP were included in this study. Reverse transcription-polymerase chain reaction and Western blot were used to detect the expression of miR-19-3p, insulin-like growth factor 1 (IGF-1), and the Akt/mTOR/p70S6K pathway. Cell cycle was defined by flow cytometric analysis. The combination of miR-19-3p and IGF-1 was revealed by luciferase assays. RESULTS The results of this study show that miR-19-3p was upregulated in the tissue of patients with POP, whereas COL-1 and IGF-1 expressions were lower in the POP group. miR-19-3p promoted excessive fibroblast autophagy and apoptosis. miR-19-3p negatively regulated the Akt/mTOR/p70S6K pathway and inhibited COL-1 secretion. Luciferase reporter assay showed that miR-19-3p regulated IGF-1 expression by direct target binding. CONCLUSIONS miR-19-3p has negative associations with the expression of Col-1. Our study highlights that miR-19-3p may affect the synthesis of Col-1 by targeting IGF-1 and that it may play an vital role in POP.
Collapse
|
8
|
Kim NR, David K, Corbeels K, Khalil R, Antonio L, Schollaert D, Deboel L, Ohlsson C, Gustafsson JÅ, Vangoitsenhoven R, Van der Schueren B, Decallonne B, Claessens F, Vanderschueren D, Dubois V. Testosterone Reduces Body Fat in Male Mice by Stimulation of Physical Activity Via Extrahypothalamic ERα Signaling. Endocrinology 2021; 162:bqab045. [PMID: 33674833 PMCID: PMC8140602 DOI: 10.1210/endocr/bqab045] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Indexed: 12/21/2022]
Abstract
Testosterone (T) reduces male fat mass, but the underlying mechanisms remain elusive, limiting its clinical relevance in hypogonadism-associated obesity. Here, we subjected chemically castrated high-fat diet-induced adult obese male mice to supplementation with T or the nonaromatizable androgen dihydrotestosterone (DHT) for 20 weeks. Both hormones increased lean mass, thereby indirectly increasing oxygen consumption and energy expenditure. In addition, T but not DHT decreased fat mass and increased ambulatory activity, indicating a role for aromatization into estrogens. Investigation of the pattern of aromatase expression in various murine tissues revealed the absence of Cyp19a1 expression in adipose tissue while high levels were observed in brain and gonads. In obese hypogonadal male mice with extrahypothalamic neuronal estrogen receptor alpha deletion (N-ERαKO), T still increased lean mass but was unable to decrease fat mass. The stimulatory effect of T on ambulatory activity was also abolished in N-ERαKO males. In conclusion, our work demonstrates that the fat-burning action of T is dependent on aromatization into estrogens and is at least partially mediated by the stimulation of physical activity via extrahypothalamic ERα signaling. In contrast, the increase in lean mass upon T supplementation is mediated through the androgen receptor and indirectly leads to an increase in energy expenditure, which might also contribute to the fat-burning effects of T.
Collapse
Affiliation(s)
- Na Ri Kim
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Karel David
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Katrien Corbeels
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Rougin Khalil
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Leen Antonio
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Dieter Schollaert
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Ludo Deboel
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 45, Sweden
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204-5056, USA
| | - Roman Vangoitsenhoven
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Bart Van der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Vanessa Dubois
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
9
|
Lau LY, Nguyen LT, Reverter A, Moore SS, Lynn A, McBride‐Kelly L, Phillips‐Rose L, Plath M, Macfarlane R, Vasudivan V, Morton L, Ardley R, Ye Y, Fortes MRS. Gene regulation could be attributed to TCF3 and other key transcription factors in the muscle of pubertal heifers. Vet Med Sci 2020; 6:695-710. [PMID: 32432381 PMCID: PMC7738712 DOI: 10.1002/vms3.278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 03/13/2020] [Accepted: 04/09/2020] [Indexed: 01/17/2023] Open
Abstract
Puberty is a whole-body event, driven by the hypothalamic integration of peripheral signals such as leptin or IGF-1. In the process of puberty, reproductive development is simultaneous to growth, including muscle growth. To enhance our understanding of muscle function related to puberty, we performed transcriptome analyses of muscle samples from six pre- and six post-pubertal Brahman heifers (Bos indicus). Our aims were to perform differential expression analyses and co-expression analyses to derive a regulatory gene network associate with puberty. As a result, we identified 431 differentially expressed (DEx) transcripts (genes and non-coding RNAs) when comparing pre- to post-pubertal average gene expression. The DEx transcripts were compared with all expressed transcripts in our samples (over 14,000 transcripts) for functional enrichment analyses. The DEx transcripts were associated with "extracellular region," "inflammatory response" and "hormone activity" (adjusted p < .05). Inflammatory response for muscle regeneration is a necessary aspect of muscle growth, which is accelerated during puberty. The term "hormone activity" may signal genes that respond to progesterone signalling in the muscle, as the presence of this hormone is an important difference between pre- and post-pubertal heifers in our experimental design. The DEx transcript with the highest average expression difference was a mitochondrial gene, ENSBTAG00000043574 that might be another important link between energy metabolism and puberty. In the derived co-expression gene network, we identified six hub genes: CDC5L, MYC, TCF3, RUNX2, ATF2 and CREB1. In the same network, 48 key regulators of DEx transcripts were identified, using a regulatory impact factor metric. The hub gene TCF3 was also a key regulator. The majority of the key regulators (22 genes) are members of the zinc finger family, which has been implicated in bovine puberty in other tissues. In conclusion, we described how puberty may affect muscle gene expression in cattle.
Collapse
Affiliation(s)
- Li Yieng Lau
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Loan T. Nguyen
- Queensland Alliance for Agriculture and Food InnovationThe University of QueenslandBrisbaneQLDAustralia
| | - Antonio Reverter
- CSIRO Agriculture and FoodQueensland Biosciences PrecinctBrisbaneQLDAustralia
| | - Stephen S. Moore
- Queensland Alliance for Agriculture and Food InnovationThe University of QueenslandBrisbaneQLDAustralia
| | - Aaron Lynn
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Liam McBride‐Kelly
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Louis Phillips‐Rose
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Mackenzie Plath
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Rhys Macfarlane
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Vanisha Vasudivan
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Lachlan Morton
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Ryan Ardley
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Yunan Ye
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
| | - Marina R. S. Fortes
- School of Chemistry and Molecular BiologyThe University of QueenslandBrisbaneQLDAustralia
- Queensland Alliance for Agriculture and Food InnovationThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
10
|
The Systemic Effects of Exercise on Regulators of Muscle and Bone in Girls and Women. Pediatr Exerc Sci 2020; 32:117-123. [PMID: 32531761 DOI: 10.1123/pes.2019-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/17/2019] [Accepted: 01/24/2020] [Indexed: 11/18/2022]
Abstract
PURPOSE To assess the systemic effects of an acute bout of moderate-intensity exercise on factors that are known to regulate muscle and bone growth in prepubertal girls and women. METHODS A total of 12 prepubertal girls (8-10 y) and 12 women (20-30 y) cycled at 60% maximal oxygen uptake for 1 hour followed by 1 hour recovery. Blood samples were collected at rest, mid-exercise, end of exercise, mid-recovery, and end of recovery. Plasma was analyzed for interleukin-6, chemokine ligand 1, fibroblast growth factor-2, total insulin growth factor-1 (IGF-1), and free IGF-1 using enzyme-linked immunosorbent assays assays. RESULTS Both groups had similar concentrations of systemic factors at baseline with the exception of free IGF-1, which was higher in girls (P = .001). Interleukin-6 response was lower in girls versus women (P = .04), with a difference of +105.1% at end of exercise (P < .001), +113.5% at mid-recovery (P = .001), and +93.2% at end of recovery (P = .02). Girls and women exhibited significant declines in chemokine ligand 1, fibroblast growth factor-2, and total IGF-1 during recovery. CONCLUSION Compared with women, an acute bout of moderate-intensity exercise in girls elicits a lower inflammatory response, suggesting that other mechanisms may be more important for driving the anabolic effects of exercise on muscle and bone in girls.
Collapse
|
11
|
Tamme R, Jürimäe J, Mäestu E, Remmel L, Purge P, Mengel E, Tillmann V. Association of Serum Testosterone at 12 Years with a Subsequent Increase in Bone Mineral Apparent Density at 18 Years: A Longitudinal Study of Boys in Puberty. Horm Res Paediatr 2020; 91:400-405. [PMID: 31505497 DOI: 10.1159/000502606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/09/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Cross-sectional studies have associated serum testosterone with bone mineral density (BMD). However, there is a shortage of prospective longitudinal studies in this domain, leaving it unclear whether changes in testosterone level precede changes in BMD. OBJECTIVES To examine the association between serum testosterone concentration at the age of 12 years and a subsequent increase in BMD by the age of 18 years. METHODS Eighty-eight boys with a mean age of 12.1 ± 0.7 (time point 1 [T1]) and 18.0 ± 0.7 (T2) were investigated. For both time points, serum testosterone was measured from venous blood samples. Total body (TB) and lumbar spine (LS) BMD and bone mineral apparent density (BMAD) were measured. As different brands of DEXA machines were used at T1 and T2, we calculated SD scores (SDS) from samples at T1 and T2 and their change (Δ). As covariates, bone age at T1 and physical activity (PA) by accelerometer at T1 and T2 were measured. RESULTS Serum testosterone at T1 was positively correlated with TB BMD at T2 (r = 0.28; p < 0.01), Δ TB BMAD SDS (r = 0.47; p < 0.0001) and Δ LS BMAD SDS (r = 0.23; p < 0.05). When additionally controlling for bone age and total PA at T1, the correlation between testosterone at T1 and Δ TB BMAD SDS remained significant (r = 0.32; p < 0.05). CONCLUSIONS Serum testosterone concentration at the age of 12 years is associated with a subsequent increase in TB BMAD by the age of 18 years. This supports the inference that testosterone levels in early puberty may influence subsequent bone mineral accrual.
Collapse
Affiliation(s)
- Reeli Tamme
- Faculty of Medicine, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia, .,Children's Clinic of Tartu University Hospital, Tartu, Estonia,
| | - Jaak Jürimäe
- Faculty of Medicine, Institute of Sports Sciences and Physiotherapy, University of Tartu, Tartu, Estonia
| | - Evelin Mäestu
- Faculty of Medicine, Institute of Sports Sciences and Physiotherapy, University of Tartu, Tartu, Estonia
| | - Liina Remmel
- Faculty of Medicine, Institute of Sports Sciences and Physiotherapy, University of Tartu, Tartu, Estonia
| | - Priit Purge
- Faculty of Medicine, Institute of Sports Sciences and Physiotherapy, University of Tartu, Tartu, Estonia
| | - Eva Mengel
- Children's Clinic of Tartu University Hospital, Tartu, Estonia
| | - Vallo Tillmann
- Faculty of Medicine, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.,Children's Clinic of Tartu University Hospital, Tartu, Estonia
| |
Collapse
|
12
|
Chen JF, Lin PW, Tsai YR, Yang YC, Kang HY. Androgens and Androgen Receptor Actions on Bone Health and Disease: From Androgen Deficiency to Androgen Therapy. Cells 2019; 8:cells8111318. [PMID: 31731497 PMCID: PMC6912771 DOI: 10.3390/cells8111318] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022] Open
Abstract
Androgens are not only essential for bone development but for the maintenance of bone mass. Therefore, conditions with androgen deficiency, such as male hypogonadism, androgen-insensitive syndromes, and prostate cancer with androgen deprivation therapy are strongly associated with bone loss and increased fracture risk. Here we summarize the skeletal effects of androgens—androgen receptors (AR) actions based on in vitro and in vivo studies from animals and humans, and discuss bone loss due to androgens/AR deficiency to clarify the molecular basis for the anabolic action of androgens and AR in bone homeostasis and unravel the functions of androgen/AR signaling in healthy and disease states. Moreover, we provide evidence for the skeletal benefits of androgen therapy and elucidate why androgens are more beneficial than male sexual hormones, highlighting their therapeutic potential as osteoanabolic steroids in improving bone fracture repair. Finally, the application of selective androgen receptor modulators may provide new approaches for the treatment of osteoporosis and fractures as well as building stronger bones in diseases dependent on androgens/AR status.
Collapse
Affiliation(s)
- Jia-Feng Chen
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Kaohsiung Chang-Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 833, Taiwan;
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan; (P.-W.L.); (Y.-R.T.); (Y.-C.Y.)
| | - Pei-Wen Lin
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan; (P.-W.L.); (Y.-R.T.); (Y.-C.Y.)
- Center for Menopause and Reproductive Medicine Research, Department of Obstetrics and Gynecology, Kaohsiung Chang-Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 833, Taiwan
| | - Yi-Ru Tsai
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan; (P.-W.L.); (Y.-R.T.); (Y.-C.Y.)
- Center for Menopause and Reproductive Medicine Research, Department of Obstetrics and Gynecology, Kaohsiung Chang-Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 833, Taiwan
- An-Ten Obstetrics and Gynecology Clinic, Kaohsiung 802, Taiwan
| | - Yi-Chien Yang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan; (P.-W.L.); (Y.-R.T.); (Y.-C.Y.)
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Hong-Yo Kang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan; (P.-W.L.); (Y.-R.T.); (Y.-C.Y.)
- Center for Menopause and Reproductive Medicine Research, Department of Obstetrics and Gynecology, Kaohsiung Chang-Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 833, Taiwan
- Correspondence: ; Tel.: +886-7-731-7123 (ext. 8898)
| |
Collapse
|
13
|
Age and sex effects on the relationship between body composition and hip geometric structure in males and females from East China. Arch Osteoporos 2018; 13:79. [PMID: 30019139 DOI: 10.1007/s11657-018-0488-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/19/2018] [Indexed: 02/03/2023]
Abstract
UNLABELLED The study finds bone mineral density is the principal determinant of hip geometry and lean mass is a better determinant than fat mass in Chinese. Moreover, the impact of fat on skeleton differs with age, with a negative effect in young people but a more positive effect in elderly. PURPOSE The aim of this study was to examine whether the correlation between body composition including bone mineral density (BMD), lean mass (LM) and fat mass (FM), and hip geometric structure change with aging in males and females from East China. METHODS It was a cross-section study. A total of 1168 healthy male and 1066 healthy females in Shanghai were divided into six groups based on their age and sex. All participants were evaluated by assessing the BMD of lumber spine and proximal hip, total LM, total FM, and geometric parameters of the hip such as the cross-sectional area (CSA), average cortical thickness (ACT), and the buckling ratio (BR) at the narrow neck (NN), the intertrochanter (IT), and the shaft (FS). RESULTS Among the three body composition, the correlation between hip BMD and hip geometric structure was strongest. LM showed significantly positive correlations with CSA. FM showed negative or little positive correlation with hip geometry in the young group. However, the degree of the contribution of FM to hip geometric structure became substantially positive with aging. Limb LM produced the largest positive contribution to CSA and ACT at all three regions in young males. However, in older males the trunk LM produced the largest positive contribution to CSA and ACT. CONCLUSIONS Among all body composition parameters, hip BMD showed the largest correlation with hip geometric structure, with LM showing the second largest. The impact of FM and LM on hip geometry changed with aging and with different distributions of lean mass and fat mass.
Collapse
|
14
|
André GI, Firman RC, Simmons LW. Phenotypic plasticity in genitalia: baculum shape responds to sperm competition risk in house mice. Proc Biol Sci 2018; 285:20181086. [PMID: 30051823 PMCID: PMC6053933 DOI: 10.1098/rspb.2018.1086] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/15/2018] [Indexed: 12/26/2022] Open
Abstract
Males are known to adjust their expenditure on testes growth and sperm production in response to sperm competition risk. Genital morphology can also contribute to competitive fertilization success but whether male genital morphology can respond plastically to the sperm competition environment has received little attention. Here, we exposed male house mice to two different sperm competition environments during their sexual development and quantified phenotypic plasticity in baculum morphology. The sperm competition environment generated plasticity in body growth. Males maturing under sperm competition risk were larger and heavier than males maturing under no sperm competition risk. We used a landmark-based geometric morphometric approach to measure baculum size and shape. Independent of variation in body size, males maintained under risk of sperm competition had a relatively thicker and more distally extended baculum bulb compared with males maintained under no sperm competition risk. Plasticity in baculum shape paralleled evolutionary responses to selection from sperm competition reported in previous studies of house mice. Our findings provide experimental evidence of socially mediated phenotypic plasticity in male genitalia.
Collapse
Affiliation(s)
- Gonçalo I André
- Centre for Evolutionary Biology, School of Biological Sciences (M092), The University of Western Australia, Crawley 6009, Australia
| | - Renée C Firman
- Centre for Evolutionary Biology, School of Biological Sciences (M092), The University of Western Australia, Crawley 6009, Australia
| | - Leigh W Simmons
- Centre for Evolutionary Biology, School of Biological Sciences (M092), The University of Western Australia, Crawley 6009, Australia
| |
Collapse
|
15
|
Sretenovic J, Ajdzanovic V, Zivkovic V, Srejovic I, Corbic M, Milosevic V, Jakovljevic V, Milosavljevic Z. Nandrolone decanoate and physical activity affect quadriceps in peripubertal rats. Acta Histochem 2018; 120:429-437. [PMID: 29759662 DOI: 10.1016/j.acthis.2018.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/27/2018] [Accepted: 04/30/2018] [Indexed: 02/06/2023]
Abstract
Anabolic androgenic steroids (AASs) are synthetic analogs of testosterone often used by athletes to increase the skeletal muscle mass. Our goal was to examine the effects of physical activity and physical activity combined with supraphysiological doses of nandrolone on functional morphology of the quadriceps muscle. The study included 32 peripubertal Wistar rats, divided into 4 groups: control (T-N-), nandrolone (T-N+), physical activity (T+N-) and physical activity plus nandrolone (T+N+) groups. The T+N- and T+N+ group swam for 4 weeks, 1 h/day, 5 days/week. The T-N+ and T+N+ groups received nandolone decanoate (20 mg/kg b.w.) once per week, subcutaneously. Subsequently, the rats were sacrificed and muscle specimens were prepared for the processing. Tissue sections were histochemically and immunohistochemically stained, while the image analysis was used for quantification. Longitudinal diameter of quadriceps muscle cells was increased for 21% in T-N+, for 57% in T+N- and for 64% in T+N+ group while cross section muscle cell area was increased in T-N+ for 19%, in T+N- for 47% and in T+N+ group for 59%, compared to the control. Collagen fibers covered area was increased in T-N+ group for 36%, in T+N- for 109% and in T+N+ group for 159%, compared to the control. Erythrocyte depots were decreased in T-N+ group and increased in T+N- and T+N+ group, in comparison with T-N-. VEGF depots were increased in all treated groups. Chronic administration of supraphysiological doses of AASs alone or in combination with physical activity induces hypertrophy and significant changes in the quadriceps muscle tissue structure.
Collapse
Affiliation(s)
- Jasmina Sretenovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Vladimir Ajdzanovic
- Department of Cytology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Vladimir Zivkovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Ivan Srejovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Milena Corbic
- Clinic of Neurology, KRH Klinikum Agnes Karll Laatzen, Hannover, Germany
| | - Verica Milosevic
- Department of Cytology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia.
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia; Department of Human Pathology, 1st Moscow State Medical University IM Sechenov, Moscow, Russia
| | - Zoran Milosavljevic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
16
|
Muscle-Bone Crosstalk: Emerging Opportunities for Novel Therapeutic Approaches to Treat Musculoskeletal Pathologies. Biomedicines 2017; 5:biomedicines5040062. [PMID: 29064421 PMCID: PMC5744086 DOI: 10.3390/biomedicines5040062] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/09/2017] [Accepted: 10/18/2017] [Indexed: 12/30/2022] Open
Abstract
Osteoporosis and sarcopenia are age-related musculoskeletal pathologies that often develop in parallel. Osteoporosis is characterized by a reduced bone mass and an increased fracture risk. Sarcopenia describes muscle wasting with an increasing risk of injuries due to falls. The medical treatment of both diseases costs billions in health care per year. With the impact on public health and economy, and considering the increasing life expectancy of populations, more efficient treatment regimens are sought. The biomechanical interaction between both tissues with muscle acting on bone is well established. Recently, both tissues were also determined as secretory endocrine organs affecting the function of one another. New exciting discoveries on this front are made each year, with novel signaling molecules being discovered and potential controversies being described. While this review does not claim completeness, it will summarize the current knowledge on both the biomechanical and the biochemical link between muscle and bone. The review will highlight the known secreted molecules by both tissues affecting the other and finish with an outlook on novel therapeutics that could emerge from these discoveries.
Collapse
|
17
|
Overexpressing the novel autocrine/endocrine adipokine WISP2 induces hyperplasia of the heart, white and brown adipose tissues and prevents insulin resistance. Sci Rep 2017; 7:43515. [PMID: 28240264 PMCID: PMC5327486 DOI: 10.1038/srep43515] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 01/27/2017] [Indexed: 12/31/2022] Open
Abstract
WISP2 is a novel adipokine, most highly expressed in the adipose tissue and primarily in undifferentiated mesenchymal cells. As a secreted protein, it is an autocrine/paracrine activator of canonical WNT signaling and, as an intracellular protein, it helps to maintain precursor cells undifferentiated. To examine effects of increased WISP2 in vivo, we generated an aP2-WISP2 transgenic (Tg) mouse. These mice had increased serum levels of WISP2, increased lean body mass and whole body energy expenditure, hyperplastic brown/white adipose tissues and larger hyperplastic hearts. Obese Tg mice remained insulin sensitive, had increased glucose uptake by adipose cells and skeletal muscle in vivo and ex vivo, increased GLUT4, increased ChREBP and markers of adipose tissue lipogenesis. Serum levels of the novel fatty acid esters of hydroxy fatty acids (FAHFAs) were increased and transplantation of Tg adipose tissue improved glucose tolerance in recipient mice supporting a role of secreted FAHFAs. The growth-promoting effect of WISP2 was shown by increased BrdU incorporation in vivo and Tg serum increased mesenchymal precursor cell proliferation in vitro. In contrast to conventional canonical WNT ligands, WISP2 expression was inhibited by BMP4 thereby allowing normal induction of adipogenesis. WISP2 is a novel secreted regulator of mesenchymal tissue cellularity.
Collapse
|
18
|
Sasanuma H, Nakata M, Parmila K, Nakae J, Yada T. PDK1-FoxO1 pathway in AgRP neurons of arcuate nucleus promotes bone formation via GHRH-GH-IGF1 axis. Mol Metab 2017; 6:428-439. [PMID: 28462077 PMCID: PMC5404105 DOI: 10.1016/j.molmet.2017.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 02/11/2017] [Indexed: 01/13/2023] Open
Abstract
Objective In the hypothalamic arcuate nucleus (ARC), orexigenic agouti-related peptide (AgRP) neurons regulate feeding behavior and energy homeostasis, functions connected to bone metabolism. The 3-phosphoinositide-dependent protein kinase-1 (PDK1) serves as a major signaling molecule particularly for leptin and insulin in AgRP neurons. We asked whether PDK1 in AGRP neurons also contributes to bone metabolism. Methods We generated AgRP neuron-specific PDK1 knockout (Agrp Pdk1−/−) mice and those with additional AgRP neuron-specific expression of transactivation-defective FoxO1 (Agrp Pdk1−/−Δ256Foxo1). Bone metabolism in KO and WT mice was analyzed by quantitative computed tomography (QCT), bone histomorphometry, measurement of plasma biomarkers, and qPCR analysis of peptides. Results In Agrp Pdk1−/− female mice aged 6 weeks, compared with Agrp Cre mice, both stature and femur length were shorter while body weight was unchanged. Cortical bone mineral density (BMD) and cancellous BMD in the femur decreased, and bone formation was delayed. Furthermore, plasma GH and IGF-1 levels were reduced in parallel with decreased mRNA expressions for GH in pituitary and GHRH in ARC. Osteoblast activity was suppressed and osteoclast activity was enhanced. These changes in stature, BMD and GH level were rescued in Agrp Pdk1−/−Δ256Foxo1 mice, suggesting that the bone abnormalities and impaired GH release were mediated by enhanced Foxo1 due to deletion of PDK1. Conclusions This study reveals a novel role of PDK1-Foxo1 pathway of AgRP neurons in controlling bone metabolism primarily via GHRH-GH-IGF-1 axis. Agrp neuron-selective Pdk1 knockout mice exhibit short stature, shortened limbs and decreased bone density in both cortical and cancellous bones. In Agrp Pdk1 knockout mice, GHRH-GH-IGF1 axis was markedly down-regulated. Retarded bone growth and reduced GH in Agrp Pdk1 knockout mice were rescued by additional expression of dominant negative FoxO1 in AgRP neurons. Pdk1-FoxO1 signaling in AgRP neurons is linked to regulation of GHRH-GH-IGF1 axis and bone metabolism.
Collapse
Affiliation(s)
- Hideyuki Sasanuma
- Department of Physiology, Division of Integrative Physiology, Jichi Medical University, 3311-1 Yakushiji, Tochigi, Shimotsuke, 329-0498, Japan; Department of Orthopaedic Surgery, Faculty of Medicine, Jichi Medical University, 3311-1 Yakushiji, Tochigi, Shimotsuke, 329-0498, Japan
| | - Masanori Nakata
- Department of Physiology, Division of Integrative Physiology, Jichi Medical University, 3311-1 Yakushiji, Tochigi, Shimotsuke, 329-0498, Japan.
| | - Kumari Parmila
- Department of Physiology, Division of Integrative Physiology, Jichi Medical University, 3311-1 Yakushiji, Tochigi, Shimotsuke, 329-0498, Japan
| | - Jun Nakae
- Frontier Medicine on Metabolic Syndrome, Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshihiko Yada
- Department of Physiology, Division of Integrative Physiology, Jichi Medical University, 3311-1 Yakushiji, Tochigi, Shimotsuke, 329-0498, Japan.
| |
Collapse
|
19
|
Combined Effects of Androgen and Growth Hormone on Osteoblast Marker Expression in Mouse C2C12 and MC3T3-E1 Cells Induced by Bone Morphogenetic Protein. J Clin Med 2017; 6:jcm6010006. [PMID: 28067796 PMCID: PMC5294959 DOI: 10.3390/jcm6010006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 12/07/2016] [Accepted: 01/03/2017] [Indexed: 11/17/2022] Open
Abstract
Osteoblasts undergo differentiation in response to various factors, including growth factors and steroids. Bone mass is diminished in androgen- and/or growth hormone (GH)-deficient patients. However the functional relationship between androgen and GH, and their combined effects on bone metabolism, remains unclear. Here we investigated the mutual effects of androgen and GH on osteoblastic marker expression using mouse myoblastic C2C12 and osteoblast-like MC3T3-E1 cells. Combined treatment with dihydrotestosterone (DHT) and GH enhanced BMP-2-induced expression of Runx2, ALP, and osteocalcin mRNA, compared with the individual treatments in C2C12 cells. Co-treatment with DHT and GH activated Smad1/5/8 phosphorylation, Id-1 transcription, and ALP activity induced by BMP-2 in C2C12 cells but not in MC3T3-E1 cells. The insulin-like growth factor (IGF-I) mRNA level was amplified by GH and BMP-2 treatment and was restored by co-treatment with DHT in C2C12 cells. The mRNA level of the IGF-I receptor was not significantly altered by GH or DHT, while it was increased by IGF-I. In addition, IGF-I treatment increased collagen-1 mRNA expression, whereas blockage of endogenous IGF-I activity using an anti-IGF-I antibody failed to suppress the effect of GH and DHT on BMP-2-induced Runx2 expression in C2C12 cells, suggesting that endogenous IGF-I was not substantially involved in the underlying GH actions. On the other hand, androgen receptor and GH receptor mRNA expression was suppressed by BMP-2 in both cell lines, implying the existence of a feedback action. Collectively the results showed that the combined effects of androgen and GH facilitated BMP-2-induced osteoblast differentiation at an early stage by upregulating BMP receptor signaling.
Collapse
|
20
|
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: 484] [Impact Index Per Article: 69.1] [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
|
21
|
Improda N, Capalbo D, Esposito A, Salerno M. Muscle and skeletal health in children and adolescents with GH deficiency. Best Pract Res Clin Endocrinol Metab 2016; 30:771-783. [PMID: 27974190 DOI: 10.1016/j.beem.2016.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In addition to promoting linear growth, GH plays a key role in the regulation of bone and muscle development and metabolism. Although GH deficiency is frequently listed among the causes of secondary osteoporosis in children, its impact on bone and muscle health and on fracture risk is still not completely established. Current data suggest that childhood-onset GH deficiency can affect bone and muscle mass and strength, with GH replacement therapy exerting beneficial effects. Moreover, GH withdrawal at final height can result in reduced peak bone and muscle mass, potentially leading to increased fracture risk in adulthood. Thus, the muscle-bone unit in GH deficient subjects should be monitored during childhood and adolescence in order to prevent osteoporosis and increased fracture risk and GH replacement should be tailored to ensure an optimal bone and muscle health.
Collapse
Affiliation(s)
- Nicola Improda
- Department of Medical Translational Sciences, Paediatric Endocrinology Section, Federico II University, Via S. Pansini 5, 80131 Naples, Italy.
| | - Donatella Capalbo
- Department of Paediatrics, Federico II University, Via S. Pansini 5, 80131 Naples, Italy.
| | - Andrea Esposito
- Department of Medical Translational Sciences, Paediatric Endocrinology Section, Federico II University, Via S. Pansini 5, 80131 Naples, Italy.
| | - Mariacarolina Salerno
- Department of Medical Translational Sciences, Paediatric Endocrinology Section, Federico II University, Via S. Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
22
|
Laurent MR, Dubois V, Claessens F, Verschueren SMP, Vanderschueren D, Gielen E, Jardí F. Muscle-bone interactions: From experimental models to the clinic? A critical update. Mol Cell Endocrinol 2016; 432:14-36. [PMID: 26506009 DOI: 10.1016/j.mce.2015.10.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/13/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023]
Abstract
Bone is a biomechanical tissue shaped by forces from muscles and gravitation. Simultaneous bone and muscle decay and dysfunction (osteosarcopenia or sarco-osteoporosis) is seen in ageing, numerous clinical situations including after stroke or paralysis, in neuromuscular dystrophies, glucocorticoid excess, or in association with vitamin D, growth hormone/insulin like growth factor or sex steroid deficiency, as well as in spaceflight. Physical exercise may be beneficial in these situations, but further work is still needed to translate acceptable and effective biomechanical interventions like vibration therapy from animal models to humans. Novel antiresorptive and anabolic therapies are emerging for osteoporosis as well as drugs for sarcopenia, cancer cachexia or muscle wasting disorders, including antibodies against myostatin or activin receptor type IIA and IIB (e.g. bimagrumab). Ideally, increasing muscle mass would increase muscle strength and restore bone loss from disuse. However, the classical view that muscle is unidirectionally dominant over bone via mechanical loading is overly simplistic. Indeed, recent studies indicate a role for neuronal regulation of not only muscle but also bone metabolism, bone signaling pathways like receptor activator of nuclear factor kappa-B ligand (RANKL) implicated in muscle biology, myokines affecting bone and possible bone-to-muscle communication. Moreover, pharmacological strategies inducing isolated myocyte hypertrophy may not translate into increased muscle power because tendons, connective tissue, neurons and energy metabolism need to adapt as well. We aim here to critically review key musculoskeletal molecular pathways involved in mechanoregulation and their effect on the bone-muscle unit as a whole, as well as preclinical and emerging clinical evidence regarding the effects of sarcopenia therapies on osteoporosis and vice versa.
Collapse
Affiliation(s)
- Michaël R Laurent
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium.
| | - Vanessa Dubois
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Frank Claessens
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Sabine M P Verschueren
- Research Group for Musculoskeletal Rehabilitation, Department of Rehabilitation Science, KU Leuven, 3000 Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Evelien Gielen
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ferran Jardí
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
23
|
Choi YY, Choi Y, Kim J, Choi H, Shin J, Roh J. Peripubertal Caffeine Exposure Impairs Longitudinal Bone Growth in Immature Male Rats in a Dose- and Time-Dependent Manner. J Med Food 2016; 19:73-84. [DOI: 10.1089/jmf.2015.3467] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Yun-Young Choi
- Department of Nuclear Medicine, College of Medicine, Hanyang University, Seoul, Korea
| | - Yuri Choi
- Laboratory of Reproductive Endocrinology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, Korea
| | - Jisook Kim
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - Hyeonhae Choi
- Laboratory of Reproductive Endocrinology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, Korea
| | - Jiwon Shin
- Laboratory of Reproductive Endocrinology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, Korea
| | - Jaesook Roh
- Laboratory of Reproductive Endocrinology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, Korea
| |
Collapse
|
24
|
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: 1.3] [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
|
25
|
Shin J, Choi Y, Kim J, Yu AR, Shin JS, Choi YY, Roh J. High doses of caffeine reduce in vivo osteogenic activity in prepubertal rats. J Anat 2015; 227:10-20. [PMID: 26041429 DOI: 10.1111/joa.12332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2015] [Indexed: 11/27/2022] Open
Abstract
Caffeine adversely affects endochondral ossification during fetal skeletal growth, and results in increased incidence of delayed and abnormal fetal skeletal development. Chronic caffeine intake also decreases growth hormone secretion. Thus, it is conceivable that caffeine may disrupt bone growth during the peripubertal period. This study aimed to investigate the impact of high-caffeine consumption on bone growth throughout puberty. A total of 51 male rats (21 days old) were divided randomly into three groups: a control group and two groups fed caffeine via gavage with 120 and 180 mg kg(-1) day(-1) for 4 weeks. After death, the final length and weight of leg bones were measured, and the tibia processed for histomorphometric analysis. Caffeine caused a significant decrease in body mass gain. This was accompanied with proportional decreases in lean body mass and body fat. In addition, bone mass and osteogenic activity in vivo were assessed using dual-energy X-ray absorptiometry and (18) F-NaF positron emission tomography. The results showed significant decreases of bone mass and in vivo osteogenic activity in the caffeine-fed groups. Rats fed with caffeine showed a significantly shorter and lighter tibia and femur and the vertebral column compared with controls. In addition, caffeine does not increase the width of the growth plates (GPs), it slows the rate at which the GP closes due to a slower rate of growth. These results demonstrated that caffeine altered osteogenic activity, leading to delayed peripubertal longitudinal bone growth and maturation. Given that osteogenic cells undergo dynamic changes in metabolic activity and that the pubertal growth spurt is mainly stimulated by growth hormone/insulin-like growth factor-1 and sex steroids during pubertal development, caffeine could suppress ossification by interfering with both physiological changes in hormonal secretion and osteogenic activity during this critical period. Further study will be needed to investigate the cellular/molecular mechanism by which caffeine affects osteogenesis using in vitro experimental models.
Collapse
Affiliation(s)
- Jiwon Shin
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul, Korea
| | - Yuri Choi
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul, Korea
| | - Jisook Kim
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - A-Ram Yu
- Department of Nuclear Medicine, College of Medicine, Hanyang University, Seoul, Korea
| | - Ji-Soo Shin
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul, Korea
| | - Yun-Young Choi
- Department of Nuclear Medicine, College of Medicine, Hanyang University, Seoul, Korea
| | - Jaesook Roh
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul, Korea
| |
Collapse
|
26
|
Russell PK, Clarke MV, Cheong K, Anderson PH, Morris HA, Wiren KM, Zajac JD, Davey RA. Androgen receptor action in osteoblasts in male mice is dependent on their stage of maturation. J Bone Miner Res 2015; 30:809-23. [PMID: 25407961 DOI: 10.1002/jbmr.2413] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 11/12/2014] [Accepted: 11/14/2014] [Indexed: 11/10/2022]
Abstract
Androgen action via the androgen receptor (AR) is essential for normal skeletal growth and bone maintenance post-puberty in males; however, the molecular and cellular mechanisms by which androgens exert their actions in osteoblasts remains relatively unexplored in vivo. To identify autonomous AR actions in osteoblasts independent of AR signaling in other tissues, we compared the extent to which the bone phenotype of the Global-ARKO mouse was restored by replacing the AR in osteoblasts commencing at either the (1) proliferative or (2) mineralization stage of their maturation. In trabecular bone, androgens stimulated trabecular bone accrual during growth via the AR in proliferating osteoblasts and maintained trabecular bone post-puberty via the AR in mineralizing osteoblasts, with its predominant action being to inhibit bone resorption by decreasing the ratio of receptor activator of NF-κB ligand (RANKL) to osteoprotegerin (OPG) gene expression. During growth, replacement of the AR in proliferating but not mineralizing osteoblasts of Global-ARKOs was able to partially restore periosteal circumference, supporting the concept that androgen action in cortical bone to increase bone size during growth is mediated via the AR in proliferating osteoblasts. This study provides further significant insight into the mechanism of androgen action via the AR in osteoblasts, demonstrating that it is dependent on the stage of osteoblast maturation.
Collapse
Affiliation(s)
- Patricia K Russell
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Australia
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Park M, Choi Y, Choi H, Yim JY, Roh J. High Doses of Caffeine during the Peripubertal Period in the Rat Impair the Growth and Function of the Testis. Int J Endocrinol 2015; 2015:368475. [PMID: 25983753 PMCID: PMC4423020 DOI: 10.1155/2015/368475] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/06/2015] [Indexed: 11/18/2022] Open
Abstract
Prenatal caffeine exposure adversely affects the development of the reproductive organs of male rat offspring. Thus, it is conceivable that peripubertal caffeine exposure would also influence physiologic gonadal changes and function during this critical period for sexual maturation. This study investigated the impact of high doses of caffeine on the testes of prepubertal male rats. A total of 45 immature male rats were divided randomly into three groups: a control group and 2 groups fed 120 and 180 mg/kg/day of caffeine, respectively, via the stomach for 4 weeks. Caffeine caused a significant decrease in body weight gain, accompanied by proportional decreases in lean body mass and body fat. The caffeine-fed animals had smaller and lighter testes than those of the control that were accompanied by negative influences on the histologic parameters of the testes. In addition, stimulated-testosterone ex vivo production was reduced in Leydig cells retrieved from the caffeine-fed animals. Our results demonstrate that peripubertal caffeine consumption can interfere with the maturation and function of the testis, possibly by interrupting endogenous testosterone secretion and reducing the sensitivity of Leydig cells to gonadotrophic stimulation. In addition, we confirmed that pubertal administration of caffeine reduced testis growth and altered testis histomorphology.
Collapse
Affiliation(s)
- Minji Park
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul 133-791, Republic of Korea
| | - Yuri Choi
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul 133-791, Republic of Korea
| | - Hyeonhae Choi
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul 133-791, Republic of Korea
| | - Ju-Yearn Yim
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul 133-791, Republic of Korea
| | - Jaesook Roh
- Laboratory of Reproductive Endocrinology, Department of Anatomy & Cell Biology, College of Medicine, Hanyang University, Seoul 133-791, Republic of Korea
- *Jaesook Roh:
| |
Collapse
|
28
|
Aghamir SMK, Salavati A, Yousefie R, Tootian Z, Ghazaleh N, Jamali M, Azimi P. Does bone marrow-derived mesenchymal stem cell transfusion prevent antisperm antibody production after traumatic testis rupture? Urology 2014; 84:82-6. [PMID: 24797037 DOI: 10.1016/j.urology.2014.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 02/10/2014] [Accepted: 03/04/2014] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To determine whether transfusion of mesenchymal stem cells (MSCs) could prevent humoral immune response and autoimmunization against sperms after traumatic testis rupture. METHODS Immunomodulatory properties of MSCs have been evaluated by a prospective cohort on 50 adult BALB/c mice. In each interventional arms of study, controlled testis rupture and surgical repair were exerted. In addition to tissue repair, single dose of 5×10(5) MSCs labeled by green fluorescent protein was delivered intravenously to 20 cases (cell therapy group). After euthanizing, seroconversion of antisperm antibody (ASA) was compared between 2 interventional groups as response of humoral immune system. Lung and testis tissues were examined for green fluorescent protein-positive cells to assess whether presence of stem cells is correlated with seroconversion rates. RESULTS Six cases had been lost during the study. Fourteen of 16 mice in cell therapy control group formed ASA (87.5%) but 6 of 18 mice (33.3%) in cell therapy group were immunized and formed ASA (P=.002). Transplanted cells were traced in lungs of 55% (n=10) of cell therapy group and none were found in trauma site. Small volume of mice blood was our main limitation to trace seroconversion or quantitative measurement of ASA in each case. CONCLUSION In this in vivo model of autoimmune infertility, bone marrow-derived MSC transfusion showed immunosuppressive effects on antibody production. Considering immunomodulatory properties of MSCs even in allogeneic settings, novel clinical application should be investigated further.
Collapse
Affiliation(s)
- Seyyed Mohammad Kazem Aghamir
- Urology Department, Tehran University of Medical Sciences, Tehran, Iran; Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Alborz Salavati
- Urology Department, Tehran University of Medical Sciences, Tehran, Iran.
| | - Reza Yousefie
- Urology Department, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | - Pourya Azimi
- Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Ultrastructure of Intervertebral Disc and Vertebra-Disc Junctions Zones as a Link in Etiopathogenesis of Idiopathic Scoliosis. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/850594] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Background Context. There is no general accepted theory on the etiology of idiopathic scoliosis (IS). An important role of the vertebrae endplate physes (VEPh) and intervertebral discs (IVD) in spinal curve progression is acknowledged, but ultrastructural mechanisms are not well understood. Purpose. To analyze the current literature on ultrastructural characteristics of VEPh and IVD in the context of IS etiology. Study Design/Setting. A literature review. Results. There is strong evidence for multifactorial etiology of IS. Early wedging of vertebra bodies is likely due to laterally directed appositional bone growth at the concave side, caused by a combination of increased cell proliferation at the vertebrae endplate and altered mechanical properties of the outer annulus fibrosus of the adjacent IVD. Genetic defects in bending proteins necessary for IVD lamellar organization underlie altered mechanical properties. Asymmetrical ligaments, muscular stretch, and spine instability may also play roles in curve formation. Conclusions. Development of a reliable, cost effective method for identifying patients at high risk for curve progression is needed and could lead to a paradigm shift in treatment options. Unnecessary anxiety, bracing, and radiation could potentially be minimized and high risk patient could receive surgery earlier, rendering better outcomes with fewer fused segments needed to mitigate curve progression.
Collapse
|
30
|
Trobec K, von Haehling S, Anker SD, Lainscak M. Growth hormone, insulin-like growth factor 1, and insulin signaling-a pharmacological target in body wasting and cachexia. J Cachexia Sarcopenia Muscle 2011; 2:191-200. [PMID: 22207907 PMCID: PMC3222822 DOI: 10.1007/s13539-011-0043-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 10/04/2011] [Indexed: 12/26/2022] Open
Abstract
Cachexia is an irreversible process that can develop in the course of chronic disease. It is characterized by the remodeling of the metabolic, inflammatory, and endocrine pathways. Insulin, growth hormone (GH), and insulin-like growth factor 1 (IGF-1) are involved in glucose, protein, and fat metabolism, which regulates body composition. In body wasting and cachexia, their signaling is impaired and causes anabolic/catabolic imbalance. Important mechanisms include inflammatory cytokines and neurohormonal activation. Remodeled post-receptor insulin, GH, and IGF-1 pathways constitute a potential target for pharmacological treatment in the setting of body wasting and cachexia. Peroxisome proliferator-activated receptor gamma agonists, drugs inhibiting angiotensin II action (angiotensin II antagonists and inhibitors of angiotensin-converting enzyme), and testosterone, which interfere with post-receptor pathways of insulin, GH, and IGF-1, were investigated as pharmacological intervention targets and various clinically important implications were reported. There are several other potential targets, but their treatment feasibility and applicability is yet to be established.
Collapse
Affiliation(s)
- Katja Trobec
- Hospital Pharmacy; University Clinic of Respiratory and Allergic Diseases Golnik; Golnik
| | - Stephan von Haehling
- Applied Cachexia Research, Department of Cardiology; Charité Medical School, Campus Virchow-Klinikum; Berlin
- Center for Cardiovascular Research (CCR); Charité Medical School, Campus Mitte; Berlin
| | - Stefan D. Anker
- Applied Cachexia Research, Department of Cardiology; Charité Medical School, Campus Virchow-Klinikum; Berlin
- Center for Clinical and Basic Research, IRCCS San Raffaele; Rome
| | - Mitja Lainscak
- Applied Cachexia Research, Department of Cardiology; Charité Medical School, Campus Virchow-Klinikum; Berlin
- Division of Cardiology; University Clinic of Respiratory and Allergic Diseases Golnik; Golnik 36 SI-4204 Golnik
| |
Collapse
|
31
|
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: 137] [Impact Index Per Article: 10.5] [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
|
32
|
Luque RM, Lin Q, Córdoba-Chacón J, Subbaiah PV, Buch T, Waisman A, Vankelecom H, Kineman RD. Metabolic impact of adult-onset, isolated, growth hormone deficiency (AOiGHD) due to destruction of pituitary somatotropes. PLoS One 2011; 6:e15767. [PMID: 21283519 PMCID: PMC3023710 DOI: 10.1371/journal.pone.0015767] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 11/26/2010] [Indexed: 01/06/2023] Open
Abstract
Growth hormone (GH) inhibits fat accumulation and promotes protein accretion, therefore the fall in GH observed with weight gain and normal aging may contribute to metabolic dysfunction. To directly test this hypothesis a novel mouse model of adult onset-isolated GH deficiency (AOiGHD) was generated by cross breeding rat GH promoter-driven Cre recombinase mice (Cre) with inducible diphtheria toxin receptor mice (iDTR) and treating adult Cre+/−,iDTR+/− offspring with DT to selectively destroy the somatotrope population of the anterior pituitary gland, leading to a reduction in circulating GH and IGF-I levels. DT-treated Cre−/−,iDTR+/− mice were used as GH-intact controls. AOiGHD improved whole body insulin sensitivity in both low-fat and high-fat fed mice. Consistent with improved insulin sensitivity, indirect calorimetry revealed AOiGHD mice preferentially utilized carbohydrates for energy metabolism, as compared to GH-intact controls. In high-fat, but not low-fat fed AOiGHD mice, fat mass increased, hepatic lipids decreased and glucose clearance and insulin output were impaired. These results suggest the age-related decline in GH helps to preserve systemic insulin sensitivity, and in the context of moderate caloric intake, prevents the deterioration in metabolic function. However, in the context of excess caloric intake, low GH leads to impaired insulin output, and thereby could contribute to the development of diabetes.
Collapse
Affiliation(s)
- Raul M. Luque
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States of America
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Qing Lin
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States of America
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - José Córdoba-Chacón
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), and CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Papasani V. Subbaiah
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Thorsten Buch
- Neuroimmunology Division, Institute of Experimental Immunology, Department of Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Ari Waisman
- Institute for Molecular Biology, University Medical Centre, University of Mainz, Mainz, Germany
| | - Hugo Vankelecom
- Laboratory of Tissue Plasticity, Department of Molecular Cell Biology, University of Leuven (K.U.Leuven), Leuven, Belgium
| | - Rhonda D. Kineman
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States of America
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
33
|
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: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [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
|
34
|
Serra C, Bhasin S, Tangherlini F, Barton ER, Ganno M, Zhang A, Shansky J, Vandenburgh HH, Travison TG, Jasuja R, Morris C. The role of GH and IGF-I in mediating anabolic effects of testosterone on androgen-responsive muscle. Endocrinology 2011; 152:193-206. [PMID: 21084444 PMCID: PMC3033058 DOI: 10.1210/en.2010-0802] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Testosterone (T) supplementation increases skeletal muscle mass, circulating GH, IGF-I, and im IGF-I expression, but the role of GH and IGF-I in mediating T's effects on the skeletal muscle remains poorly understood. Here, we show that T administration increased body weight and the mass of the androgen-dependent levator ani muscle in hypophysectomized as well as castrated plus hypophysectomized adult male rats. T stimulated the proliferation of primary human skeletal muscle cells (hSKMCs) in vitro, an effect blocked by transfecting hSKMCs with small interference RNA targeting human IGF-I receptor (IGF-IR). In differentiation conditions, T promoted the fusion of hSKMCs into larger myotubes, an effect attenuated by small interference RNA targeting human IGF-IR. Notably, MKR mice, which express a dominant negative form of the IGF-IR in skeletal muscle fibers, treated with a GnRH antagonist (acyline) to suppress endogenous T, responded to T administration by an attenuated increase in the levator ani muscle mass. In conclusion, circulating GH and IGF-I are not essential for mediating T's effects on an androgen-responsive skeletal muscle. IGF-I signaling plays an important role in mediating T's effects on skeletal muscle progenitor cell growth and differentiation in vitro. However, IGF-IR signaling in skeletal muscle fibers does not appear to be obligatory for mediating the anabolic effects of T on the mass of androgen-responsive skeletal muscles in mice.
Collapse
Affiliation(s)
- Carlo Serra
- Section of Endocrinology, Diabetes, and Nutrition, Boston Medical Center, 670 Albany Street, Boston, Massachusetts 02118, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Liu PY, Kalak R, Lue Y, Jia Y, Erkkila K, Zhou H, Seibel MJ, Wang C, Swerdloff RS, Dunstan CR. Genetic and hormonal control of bone volume, architecture, and remodeling in XXY mice. J Bone Miner Res 2010; 25:2148-54. [PMID: 20499350 PMCID: PMC3153317 DOI: 10.1002/jbmr.104] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Klinefelter syndrome is the most common chromosomal aneuploidy in men (XXY karyotype, 1 in 600 live births) and results in testicular (infertility and androgen deficiency) and nontesticular (cognitive impairment and osteoporosis) deficits. The extent to which skeletal changes are due to testosterone deficiency or arise directly from gene overdosage cannot be determined easily in humans. To answer this, we generated XXY mice through a four-generation breeding scheme. Eight intact XXY and 9 XY littermate controls and 8 castrated XXY mice and 8 castrated XY littermate controls were euthanized at 1 year of age. Castration occurred 6 months prior to killing. A third group of 9 XXY and 11 XY littermates were castrated and simultaneously implanted with a 1-cm Silastic testosterone capsule 8 weeks prior to sacrifice. Tibias were harvested from all three groups and examined by micro-computed tomography and histomorphometry. Blood testosterone concentration was assayed by radioimmunoassay. Compared with intact XY controls, intact androgen-deficient XXY mice had lower bone volume (6.8% +/- 1.2% versus 8.8% +/- 1.7%, mean +/- SD, p = .01) and thinner trabeculae (50 +/- 4 µm versus 57 +/- 5 µm, p = .007). Trabecular separation (270 +/- 20 µm versus 270 +/- 20 µm) or osteoclast number relative to bone surface (2.4 +/- 1.0/mm2 versus 2.7 +/- 1.5/mm2) did not differ significantly. Testosterone-replaced XXY mice continued to show lower bone volume (5.5% +/- 2.4% versus 8.1% +/- 3.5%, p = .026). They also exhibited greater trabecular separation (380 +/- 69 µm versus 324 +/- 62 µm, p = .040) but equivalent blood testosterone concentrations (6.3 +/- 1.8 ng/mL versus 8.2 +/- 4.2 ng/mL, p = .28) compared with testosterone-replaced XY littermates. In contrast, castration alone drastically decreased bone volume (p < .001), trabecular thickness (p = .05), and trabecular separation (p < .01) to such a great extent that differences between XXY and XY mice were undetectable. In conclusion, XXY mice replicate many features of human Klinefelter syndrome and therefore are a useful model for studying bone. Testosterone deficiency does not explain the bone phenotype because testosterone-replaced XXY mice show reduced bone volume despite similar blood testosterone levels.
Collapse
Affiliation(s)
- Peter Y Liu
- Division of Endocrinology, Department of Medicine, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute (LA BioMed) at Harbor-UCLA Medical Center, Torrance, CA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zengin A, Zhang L, Herzog H, Baldock PA, Sainsbury A. Neuropeptide Y and sex hormone interactions in humoral and neuronal regulation of bone and fat. Trends Endocrinol Metab 2010; 21:411-8. [PMID: 20202858 DOI: 10.1016/j.tem.2010.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Revised: 02/09/2010] [Accepted: 02/09/2010] [Indexed: 02/03/2023]
Abstract
The hypothalamus regulates the skeleton and adipose tissue via endocrine mechanisms. Changes in sex steroid levels in menopause and aging are central to the associated changes in bone mass and adiposity. Whereas many of these effects occur via direct actions on osteoblasts or adipocytes, sex hormones can also mediate effects on bone and adipose tissue via interaction with neuronal pathways. A key hypothalamic regulator of bone and adipose tissue is neuropeptide Y (NPY), which coordinately influences these tissues via effects on neuroendocrine and sympathetic nervous output. Better understanding of the interaction between NPY and sex steroids in regulating skeletal and energy homeostasis could lead to more effective treatments for osteoporosis and obesity.
Collapse
Affiliation(s)
- Ayse Zengin
- Osteoporosis and Bone Biology Program, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst, Sydney, New South Wales (NSW), Australia
| | | | | | | | | |
Collapse
|
37
|
Dillon EL, Durham WJ, Urban RJ, Sheffield-Moore M. Hormone treatment and muscle anabolism during aging: androgens. Clin Nutr 2010; 29:697-700. [PMID: 20452103 DOI: 10.1016/j.clnu.2010.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 02/11/2010] [Accepted: 03/27/2010] [Indexed: 01/27/2023]
Abstract
Aging is associated with a gradual decline in circulating testosterone concentrations and decreased musculature in men. While testosterone administration is often considered when symptoms of hypogonadism are presented, the long-term effects of androgen use on muscle physiology are not yet fully understood. The definition of hypogonadism in men remains obscure but is generally indicated by total testosterone concentrations less than a threshold value of 300-500 ng/dL. Androgen replacement therapy is generally safe in men and women with low endogenous testosterone concentrations. The development of selective androgen receptor modulators (SARMs) may provide additional options in treatment of hypogonadism while lowering the potential of side effects often associated with long-term androgen use. Androgen administration, either alone or in combination with other treatments, can be successful in improving muscle mass by increasing protein anabolism and reducing protein catabolism in men and women. Further research is necessary to optimize the anabolic and anticatabolic properties of androgens for treatment and prevention of muscle loss in men and women.
Collapse
Affiliation(s)
- E Lichar Dillon
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | | |
Collapse
|
38
|
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: 98] [Impact Index Per Article: 7.0] [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
|
39
|
Ashitani JI, Matsumoto N, Nakazato M. Effect of octanoic acid-rich formula on plasma ghrelin levels in cachectic patients with chronic respiratory disease. Nutr J 2009; 8:25. [PMID: 19527531 PMCID: PMC2708182 DOI: 10.1186/1475-2891-8-25] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Accepted: 06/16/2009] [Indexed: 12/11/2022] Open
Abstract
Background For cachectic patients with chronic respiratory disease (CRD), conventional enteral nutrition formula is an optional treatment to maintain energy balance. The molecular mechanisms by which enteral nutrition formula controls appetite and weight remain unknown. We examined whether enteral nutrition formula rich in octanoic acids would increase plasma levels of ghrelin, an appetite-stimulating hormone produced in the stomach, in cachectic patients with CRD. Methods Plasma ghrelin profiles in cachectic patients with CRD were assessed and compared with those in age- and sex-matched controls. Plasma levels of acyl-ghrelin, an active ghrelin modified by octanoic acids, and desacyl-ghrelin were measured separately. We examined changes in 24-h plasma ghrelin profiles before and after single administration of the formula. We also evaluated the effects of 2-week administration of the formula on plasma ghrelin levels and nutritional status in patients. Results The ratio of acyl-ghrelin to desacyl-ghrelin in plasma was lower in patients than in controls. Single administration of the formula did not change plasma desacyl-ghrelin levels, but induced an increase in acyl-ghrelin levels. Two-week treatment with the formula was effective in increasing weight and acyl-ghrelin, along with improving nutritional status in patients. Conclusion These results show that the formula contributes to increased weight, which may be associated with induction of acyl-ghrelin production in cachectic patients with CRD.
Collapse
Affiliation(s)
- Jun-Ichi Ashitani
- Third Department of Internal Medicine, Miyazaki University School of Medicine, Kihara 5200, Miyazaki 889-1692, Japan.
| | | | | |
Collapse
|
40
|
Brown D, Hikim APS, Kovacheva EL, Sinha-Hikim I. Mouse model of testosterone-induced muscle fiber hypertrophy: involvement of p38 mitogen-activated protein kinase-mediated Notch signaling. J Endocrinol 2009; 201:129-39. [PMID: 19144735 PMCID: PMC4732720 DOI: 10.1677/joe-08-0476] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
As a prerequisite for studies using mutant mice, we established a mouse model for investigating the molecular mechanisms by which testosterone (T) promotes muscle growth. Groups of six adult male mice (C57BL/6) received one of the following treatments: 1) vehicle (sterile distilled water; normal control) and 2) GnRH antagonist with empty (sham control) or 2 cm T- filled implant. Mice were killed 2, 6, and 8 weeks after treatment. T treatment for 8 weeks resulted in a significant (P<0.001) increase in fiber area of gastrocnemius muscles. T-induced fiber-hypertrophy was accompanied by up-regulation of the Notch ligand Delta 1 and activation of Notch signaling, as evidenced by increase in activated forms of Notch 1 and Notch 2. Consistent with this, we also observed an increase in the number of proliferating cell nuclear antigen (PCNA)-positive nuclei in muscles of T-treated mice, indicating that activation of Notch signaling enhanced cell proliferation. T supplementation not only triggered p38 mitogen-activated protein kinase (MAPK) activation but also concurrently inhibited c-Jun NH(2)-terminal kinase (JNK) activation within 2 weeks of treatment. Concomitant administration of SB203580, a p38 MAPK inhibitor, effectively blocked T-induced activation of Notch signaling and significantly (P<0.001) suppressed PCNA levels. Together, our results indicate that T induces muscle fiber hypertrophy through activation of Notch signaling and the inactivation of JNK together with the activation of p38 MAPK may be critical for T-induced activation of Notch signaling and, as a consequence, muscle fiber hypertrophy.
Collapse
Affiliation(s)
- Danielle Brown
- Division of Endocrinology, Charles Drew University, Los Angeles, California 90059, USA
| | | | | | | |
Collapse
|
41
|
Bartos L, Schams D, Bubenik GA. Testosterone, but not IGF-1, LH, prolactin or cortisol, may serve as antler-stimulating hormone in red deer stags (Cervus elaphus). Bone 2009; 44:691-8. [PMID: 19124089 DOI: 10.1016/j.bone.2008.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Revised: 11/12/2008] [Accepted: 12/05/2008] [Indexed: 11/28/2022]
Abstract
The role of androgens and insulin-like growth factor 1 (IGF-1) in antler growth has been disputed. We predicted that the secretory of IGF-1 may be associated with an acceleration of body growth rather than with antler growth. Furthermore we anticipated a relationship between the increase of testosterone and the progress of antler growth. If IGF-1 is involved in the stimulation of antler growth, this should be more obvious in young than in mature stags. Eight two-year-old red deer stags (Cervus elaphus), and twelve adult red deer stags were blood sampled and the length of their velvet antlers was measured in one-week intervals during the period of antler growth. Concentrations of testosterone, cortisol, IGF-1, luteinizing hormone (LH), and prolactin were determined in plasma by enzyme immunoassay or radioimmunoassay. Antler growth per day was primarily dependent on changes in testosterone concentration per day in both groups of stags. As expected, only in two-year-old stags we detected a possible role of IGF-1 in the antler growth regulation, but that was not in agreement with previously published studies. Nevertheless, this effect was still utilized in interaction with testosterone. In addition to total antler length, only concentrations of testosterone and LH were significantly higher in adult males in comparison to two-year-old males. Our present results lead us to conclude that it is not IGF-1 but testosterone which is responsible for the intensity of antler growth in subadult and adult red deer stags.
Collapse
Affiliation(s)
- Ludek Bartos
- Department of Ethology, Institute of Animal Science, Prátelství 815, 106 00 Praha 10-Uhríneves, Czech Republic.
| | | | | |
Collapse
|
42
|
Stevenson AE, Evans BAJ, Gevers EF, Elford C, McLeod RWJ, Perry MJ, El-Kasti MM, Coschigano KT, Kopchick JJ, Evans SL, Wells T. Does adiposity status influence femoral cortical strength in rodent models of growth hormone deficiency? Am J Physiol Endocrinol Metab 2009; 296:E147-56. [PMID: 19001545 PMCID: PMC2636985 DOI: 10.1152/ajpendo.90689.2008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Growth hormone (GH)-deficiency is usually associated with elevated adiposity, hyperleptinemia, and increased fracture risk. Since leptin is thought to enhance cortical bone formation, we have investigated the contribution of elevated adiposity and hyperleptinemia on femoral strength in rodent models of GH deficiency. Quantification of the transpubertal development of femoral strength in the moderately GH-deficient/hyperleptinemic Tgr rat and the profoundly GH-deficient/hypoleptinemic dw/dw rat revealed that the mechanical properties of cortical bone in these two models were similarly compromised, a 25-30% reduction in failure load being entirely due to impairment of geometric variables. In contrast, murine models of partial (GH antagonist transgenic) and complete (GH receptor-null) loss of GH signaling and elevated adiposity showed an impairment of femoral cortical strength proportionate to the reduction of GH signaling. To determine whether impaired femoral strength is exacerbated by obesity/hyperleptinemia, femoral strength was assessed in dw/dw rats following two developmental manipulations that elevate abdominal adiposity and circulating leptin, neonatal monosodium glutamate (MSG) treatment, and maintenance on an elevated fat diet. The additional impairment of femoral strength following MSG treatment is likely to have resulted from a reduction in residual activity of the hypothalamo-pituitary-GH-IGF-I axis, but consumption of elevated dietary fat, which did not reduce circulating IGF-I, failed to exacerbate the compromised femoral strength in dw/dw rats. Taken together, our data indicate that the obesity and hyperleptinemia usually associated with GH deficiency do not exert a significant influence over the strength of cortical bone.
Collapse
Affiliation(s)
- A E Stevenson
- School of Biosciences, Cardiff University, Museum Ave., Cardiff, CF10 3AX, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Berryman DE, Christiansen JS, Johannsson G, Thorner MO, Kopchick JJ. Role of the GH/IGF-1 axis in lifespan and healthspan: lessons from animal models. Growth Horm IGF Res 2008; 18:455-471. [PMID: 18710818 PMCID: PMC2631405 DOI: 10.1016/j.ghir.2008.05.005] [Citation(s) in RCA: 195] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Accepted: 05/02/2008] [Indexed: 12/18/2022]
Abstract
Animal models are fundamentally important in our quest to understand the genetic, epigenetic, and environmental factors that contribute to human aging. In comparison to humans, relatively short-lived mammals are useful models as they allow for rapid assessment of both genetic manipulation and environmental intervention as related to longevity. These models also allow for the study of clinically relevant pathologies as a function of aging. Data associated with more distant species offers additional insight and critical consideration of the basic physiological processes and molecular mechanisms that influence lifespan. Consistently, two interventions, caloric restriction and repression of the growth hormone (GH)/insulin-like growth factor-1/insulin axis, have been shown to increase lifespan in both invertebrates and vertebrate animal model systems. Caloric restriction (CR) is a nutrition intervention that robustly extends lifespan whether it is started early or later in life. Likewise, genes involved in the GH/IGF-1 signaling pathways can lengthen lifespan in vertebrates and invertebrates, implying evolutionary conservation of the molecular mechanisms. Specifically, insulin and insulin-like growth factor-1 (IGF-1)-like signaling and its downstream intracellular signaling molecules have been shown to be associated with lifespan in fruit flies and nematodes. More recently, mammalian models with reduced growth hormone (GH) and/or IGF-1 signaling have also been shown to have extended lifespans as compared to control siblings. Importantly, this research has also shown that these genetic alterations can keep the animals healthy and disease-free for longer periods and can alleviate specific age-related pathologies similar to what is observed for CR individuals. Thus, these mutations may not only extend lifespan but may also improve healthspan, the general health and quality of life of an organism as it ages. In this review, we will provide an overview of how the manipulation of the GH/IGF axis influences lifespan, highlight the invertebrate and vertebrate animal models with altered lifespan due to modifications to the GH/IGF-1 signaling cascade or homologous pathways, and discuss the basic phenotypic characteristics and healthspan of these models.
Collapse
Affiliation(s)
- Darlene E. Berryman
- School of Human and Consumer Sciences, College of Health and Human Services, Ohio University, Athens, OH 45701
| | - Jens Sandahl Christiansen
- Jens Sandahl Christiansen, Department of Endocrinology, Aarhus University Hospital, Kommunehospitalet, DK 8000 Aarhus, Denmark
| | - Gudmundur Johannsson
- Gudmundur Johannsson, MD, Research Centre for Endocrinology and Metabolism, Sahlgrenska University Hospital, S-413 45 Göteborg, Sweden
| | - Michael O. Thorner
- Michael O. Thorner, University of Virginia Health System, Endocrinology and Metabolism, Charlottesville, VA 22908
| | - John J. Kopchick
- Edison Biotechnology Institute and Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, OH 45701; Phone: (740)593-4534; Fax: (740)593-4795
| |
Collapse
|
44
|
Pampusch MS, White ME, Hathaway MR, Baxa TJ, Chung KY, Parr SL, Johnson BJ, Weber WJ, Dayton WR. Effects of implants of trenbolone acetate, estradiol, or both, on muscle insulin-like growth factor-I, insulin-like growth factor-I receptor, estrogen receptor-α, and androgen receptor messenger ribonucleic acid levels in feedlot steers1. J Anim Sci 2008; 86:3418-23. [DOI: 10.2527/jas.2008-1085] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
45
|
Abstract
The mammalian insulin-like growth factor 1 (IGF1), which is a member of a major growth-promoting signaling system, is produced by many tissues and functions throughout embryonic and postnatal development in an autocrine/paracrine fashion. In addition to this local action, IGF1 secreted by the liver and circulating in the plasma presumably acts systemically as a classical hormone. However, an endocrine role of IGF1 in growth control was disputed on the basis of the results of a conditional, liver-specific Igf1 gene knockout in mice, which reduced significantly the level of serum IGF1, but did not affect average body weight. Because alternate interpretations of these negative data were tenable, we addressed genetically the question of hormonal IGF1 action by using a positive experimental strategy based on the features of the cre/loxP recombination system. Thus, we generated bitransgenic mice carrying in an Igf1 null background a dormant Igf1 cDNA placed downstream of a transcriptional "stop" DNA sequence flanked by loxP sites (floxed) and also a cre transgene driven by a liver-specific promoter. The Igf1 cDNA, which was inserted by knock-in into the mutated and inactive Igf1 locus itself to ensure proper transcriptional regulation, was conditionally expressed from cognate promoters exclusively in the liver after Cre-mediated excision of the floxed block. Our genetic study demonstrated that the endocrine IGF1 plays a very significant role in mouse growth, as its action contributes approximately30% of the adult body size and sustains postnatal development, including the reproductive functions of both mouse sexes.
Collapse
|
46
|
Abstract
Bone size, shape and internal architecture, and not just bone mass, account for differences in bone strength between individuals, sexes and races. The differences in bone morphology in old age - whether an individual's bone size and mass occupy the 5th, 50th or 95th percentile - is determined early in life. Bone traits track from the position established early in life. Genetic and environmental factors establish the morphological features of bone through the cellular machinery of bone modelling and remodelling which adapts bone to its loading circumstance by modifying its size and shape and the distribution of its mass. The need for both strength for loading and lightness for mobility are achieved by deposition of bone where it is needed and removal of bone from where it is not. The machinery has enormous capacity during growth, as can be seen in the bone structure of the elite athlete, but not during advancing age because of changes in the cellular machinery itself and in systemic hormonal regulatory factors.
Collapse
Affiliation(s)
- Qingju Wang
- Endocrine Centre, Centaur Building, Heidelberg Repatriation Hospital/Austin Health, Heidelberg, Victoria, Australia.
| | | |
Collapse
|
47
|
Wiren KM, Semirale AA, Zhang XW, Woo A, Tommasini SM, Price C, Schaffler MB, Jepsen KJ. Targeting of androgen receptor in bone reveals a lack of androgen anabolic action and inhibition of osteogenesis: a model for compartment-specific androgen action in the skeleton. Bone 2008; 43:440-51. [PMID: 18595795 PMCID: PMC2574646 DOI: 10.1016/j.bone.2008.04.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2008] [Revised: 04/16/2008] [Accepted: 04/23/2008] [Indexed: 10/22/2022]
Abstract
Androgens are anabolic hormones that affect many tissues, including bone. However, an anabolic effect of androgen treatment on bone in eugonadal subjects has not been observed and clinical trials have been disappointing. The androgen receptor (AR) mediates biological responses to androgens. In bone tissue, both AR and the estrogen receptor (ER) are expressed. Since androgens can be converted into estrogen, the specific role of the AR in maintenance of skeletal homoeostasis remains controversial. The goal of this study was to use skeletally targeted overexpression of AR in differentiated osteoblasts as a means of elucidating the specific role(s) for AR transactivation in the mature bone compartment. Transgenic mice overexpressing AR under the control of the 2.3-kb alpha1(I)-collagen promoter fragment showed no difference in body composition, testosterone, or 17ss-estradiol levels. However, transgenic males have reduced serum osteocalcin, CTx and TRAPC5b levels, and a bone phenotype was observed. In cortical bone, high-resolution micro-computed tomography revealed no difference in periosteal perimeter but a significant reduction in cortical bone area due to an enlarged marrow cavity. Endocortical bone formation rate was also significantly inhibited. Biomechanical analyses showed decreased whole bone strength and quality, with significant reductions in all parameters tested. Trabecular morphology was altered, with increased bone volume comprised of more trabeculae that were closer together but not thicker. Expression of genes involved in bone formation and bone resorption was significantly reduced. The consequences of androgen action are compartment-specific; anabolic effects are exhibited exclusively at periosteal surfaces, but in mature osteoblasts androgens inhibited osteogenesis with detrimental effects on matrix quality, bone fragility and whole bone strength. Thus, the present data demonstrate that enhanced androgen signaling targeted to bone results in low bone turnover and inhibition of bone formation by differentiated osteoblasts. These results indicate that direct androgen action in mature osteoblasts is not anabolic, and raise concerns regarding anabolic steroid abuse in the developing skeleton or high-dose treatment in eugonadal adults.
Collapse
Affiliation(s)
- Kristine M Wiren
- Bone and Mineral Research Unit, Portland Veterans Affairs Medical Center, Portland, Oregon, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Dopamine receptor genetic polymorphisms and body composition in undernourished pastoralists: an exploration of nutrition indices among nomadic and recently settled Ariaal men of northern Kenya. BMC Evol Biol 2008; 8:173. [PMID: 18544160 PMCID: PMC2440754 DOI: 10.1186/1471-2148-8-173] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Accepted: 06/10/2008] [Indexed: 11/10/2022] Open
Abstract
Background Minor alleles of the human dopamine receptor polymorphisms, DRD2/TaqI A and DRD4/48 bp, are related to decreased functioning and/or numbers of their respective receptors and have been shown to be correlated with body mass, height and food craving. In addition, the 7R minor allele of the DRD4 gene is at a higher frequency in nomadic compared to sedentary populations. Here we examine polymorphisms in the DRD2 and DRD4 genes with respect to body mass index (BMI) and height among men in two populations of Ariaal pastoralists, one recently settled (n = 87) and the other still nomadic (n = 65). The Ariaal live in northern Kenya, are chronically undernourished and are divided socially among age-sets. Results Frequencies of the DRD4/7R and DRD2/A1 alleles were 19.4% and 28.2%, respectively and did not differ between the nomadic and settled populations. BMI was higher in those with one or two DRD4/7R alleles in the nomadic population, but lower among the settled. Post-hoc analysis suggests that the DRD4 differences in BMI were due primarily to differences in fat free body mass. Height was unrelated to either DRD2/TaqI A or DRD4/48 bp genotypes. Conclusion Our results indicate that the DRD4/7R allele may be more advantageous among nomadic than settled Ariaal men. This result suggests that a selective advantage mediated through behaviour may be responsible for the higher frequency of the 7R alleles in nomadic relative to sedentary populations around the world. In contrast to previous work, we did not find an association between DRD2 genotypes and height. Our results support the idea that human phenotypic expression of genotypes should be rigorously evaluated in diverse environments and genetic backgrounds.
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW The present review will focus on the most important recent findings with respect to skeletal androgen action. Many studies have indicated that part of the androgen action may be related to the conversion of androgens into estrogens. Therefore, some of the most recent findings of skeletal estrogen action relevant for male skeletal physiology will also be discussed. RECENT FINDINGS Androgens and estrogens stimulate bone formation and inhibit bone resorption. Sex steroids may interact with different receptors, target cells and other bone anabolic pathways. Androgen receptor and estrogen receptor signalling appear to be important for male bone formation during growth. Sex steroid signalling may involve genomic and nongenomic pathways, interaction with mechanical loading, the growth hormone/insulin-like growth factor-I axis and/or other bone anabolic pathways. Estrogen receptor alpha in osteoclasts appears to regulate bone resorption in women but not men, whereas androgen receptor signalling in osteoblasts may only partly regulate bone resorption in males. SUMMARY The latest developments indicate that androgens and estrogens are important for male bone metabolism and homeostasis and therefore selective estrogen receptor alpha and androgen receptor signalling remain interesting drug targets for the stimulation of bone formation and male skeletal integrity.
Collapse
Affiliation(s)
- Dirk Vanderschueren
- Bone Research Unit, Laboratory for Experimental Medicine and Endocrinology, Department of Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.
| | | | | | | |
Collapse
|
50
|
Yingling VR, Taylor G. Delayed pubertal development by hypothalamic suppression causes an increase in periosteal modeling but a reduction in bone strength in growing female rats. Bone 2008; 42:1137-43. [PMID: 18406225 PMCID: PMC2494865 DOI: 10.1016/j.bone.2008.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 01/28/2008] [Accepted: 02/05/2008] [Indexed: 11/23/2022]
Abstract
The timing of the pubertal growth is a critical event in skeletal development. A delay in the onset of puberty has been correlated with increased stress fracture incidence in young women and as a result, suboptimal skeletal development may affect long-term bone strength. Gonadotropin releasing hormone antagonist (GnRH-a) injections were used to delay the onset of puberty in growing female rats. 23-day-old female rats were injected with a GnRH-antagonist at 2 dosage levels (n=15/group). The Low Dose group (1.25 mg/kg/dose) received daily injections for 27 days (sacrifice 49 days). The High Dose group received (5.0 mg/kg/dose) only 5 days per week over a 26 day period (sacrifice 48 days). Calcein injections measured bone formation activity on the periosteal and endocortical surfaces. Standard histomorphometric and biomechanical analyses were performed on the femora and ash content was measured on the tibiae of all animals. Serum estradiol and insulin-like growth factor (IGF)-1 levels were assayed. Significant delays in pubertal development occurred in the two GnRH-a groups as evidenced by delayed vaginal openings, decreased uterine and ovarian weights and suppressed estradiol levels compared to control. Femoral lengths were significantly shorter in the experimental groups and serum IGF-1 levels were higher than control. Bone strength and stiffness were significantly lower in the GnRH-a groups. Cortical bone area was decreased and total area was not different between groups. There was a significant decrease in % Ct.Ar/T.Ar. The decreased bone strength may have resulted from a decrease in the amount and distribution of bone, however, stress and Young's modulus were also decreased. There was a different response between endocortical formation indices and periosteal formation indices to the GnRH-a protocol. Endocortical bone formation rates decreased and there was an increase in periosteal labeled surface. A dose response between bone strength and GnRH-a dosage was found. The data suggest that hypothalamic suppression during pubertal development resulted in decreased bone strength which may result in fracture development.
Collapse
Affiliation(s)
- Vanessa R Yingling
- Department of Kinesiology, College of Health Professions, Temple University, 1800 North Broad Street, Philadelphia, PA 19122, USA.
| | | |
Collapse
|