1
|
Gangitano E, Curreli MI, Gandini O, Masi D, Spoltore ME, Gnessi L, Lubrano C. Bone Quality Indices Correlate with Growth Hormone Secretory Capacity in Women Affected by Weight Excess: A Cross-Sectional Study. J Clin Med 2024; 13:5064. [PMID: 39274277 PMCID: PMC11395994 DOI: 10.3390/jcm13175064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/16/2024] Open
Abstract
Background/Objectives: Obesity can be associated with impaired growth hormone (GH) secretion, with possible negative repercussions on bone health. We aimed to investigate the relationships between GH secretory capacity, evaluated with GHRH + arginine stimulation test, and bone parameters, assessed with a dual-energy X-ray absorptiometer, in a population of adult female patients affected by overweight and obesity. Methods: We assessed 276 women affected by overweight or obesity referred to the High-Specialization Center for the Care of Obesity, Umberto I Polyclinic, between 2014 and 2019 with signs or symptoms of growth hormone deficiency (GHD). Results: A total of 97 patients were diagnosed with GHD, and 179 patients with normal GH secretion were considered our control group. GHD patients showed a significantly reduced trabecular bone score (TBS) (p = 0.01). Bone quality parameters corrected for body mass index (BMI) had a positive and significant linear correlation with stimulated GH secretory capacity. Conclusions: In conclusion, bone quality, evaluated by TBS and hip structural analysis, correlates with GH-stimulated secretory capacity. GHD may act as an additive factor in the alteration of bone microarchitecture in patients affected by obesity, who are already at a higher risk of fractures.
Collapse
Affiliation(s)
- Elena Gangitano
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | | | - Orietta Gandini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Davide Masi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Elena Spoltore
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Lucio Gnessi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Carla Lubrano
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
2
|
Liu L, Luo P, Wen P, Xu P. The role of magnesium in the pathogenesis of osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1406248. [PMID: 38904051 PMCID: PMC11186994 DOI: 10.3389/fendo.2024.1406248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Magnesium (Mg), a nutritional element which is essential for bone development and mineralization, has a role in the progression of osteoporosis. Osteoporosis is a multifactorial disease characterized by significant deterioration of bone microstructure and bone loss. Mg deficiency can affect bone structure in an indirect way through the two main regulators of calcium homeostasis (parathyroid hormone and vitamin D). In human osteoblasts (OBs), parathyroid hormone regulates the expression of receptor activator of nuclear factor-κ B ligand (RANKL) and osteoprotegerin (OPG) to affect osteoclast (OC) formation. In addition, Mg may also affect the vitamin D3 -mediated bone remodeling activity. vitamin D3 usually coordinates the activation of the OB and OC. The unbalanced activation OC leads to bone resorption. The RANK/RANKL/OPG axis is considered to be a key factor in the molecular mechanism of osteoporosis. Mg participates in the pathogenesis of osteoporosis by affecting the regulation of parathyroid hormone and vitamin D levels to affect the RANK/RANKL/OPG axis. Different factors affecting the axis and enhancing OC function led to bone loss and bone tissue microstructure damage, which leads to the occurrence of osteoporosis. Clinical research has shown that Mg supplementation can alleviate the symptoms of osteoporosis to some extent.
Collapse
Affiliation(s)
- Lin Liu
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Pan Luo
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pengfei Wen
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
3
|
Zhang S, Hao W, Chen D, Chen S, Li Z, Zhong F, Wang H, Wang J, Zheng Z, Zhan Z, Dai G, Liu H. Intermittent administration of PTH for the treatment of inflammatory bone loss does not enhance entheseal pathological new bone formation. Biochem Biophys Res Commun 2024; 711:149888. [PMID: 38603833 DOI: 10.1016/j.bbrc.2024.149888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/21/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
OBJECTIVE To investigate the effect of intermittent parathyroid hormone (iPTH) administration on pathological new bone formation during treatment of ankylosing spondylitis-related osteoporosis. METHODS Animal models with pathological bone formation caused by hypothetical AS pathogenesis received treatment with iPTH. We determined the effects of iPTH on bone loss and the formation of pathological new bone with micro-computed tomography (micro-CT) and histological examination. In addition, the tamoxifen-inducible conditional knockout mice (CAGGCre-ERTM; PTHflox/flox, PTH-/-) was established to delete PTH and investigate the effect of endogenous PTH on pathological new bone formation. RESULTS iPTH treatment significantly improved trabecular bone mass in the modified collagen-induced arthritis (m-CIA) model and unbalanced mechanical loading models. Meanwhile, iPTH treatment did not enhance pathological new bone formation in all types of animal models. Endogenous PTH deficiency had no effects on pathological new bone formation in unbalanced mechanical loading models. CONCLUSION Experimental animal models of AS treated with iPTH show improvement in trabecular bone density, but not entheseal pathological bone formation,indicating it may be a potential treatment for inflammatory bone loss does in AS.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Wenjun Hao
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Dongying Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Siwen Chen
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Zihao Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Fangling Zhong
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Haitao Wang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Jianru Wang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Zhaomin Zheng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China
| | - Zhongping Zhan
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Guo Dai
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China.
| | - Hui Liu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
4
|
Tang L, Zhang YY, Liu WJ, Fu Q, Zhao J, Liu YB. DNA methylation of promoter region inhibits galectin-1 expression in BMSCs of aged mice. Am J Physiol Cell Physiol 2024; 326:C429-C441. [PMID: 38105757 DOI: 10.1152/ajpcell.00334.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/01/2023] [Accepted: 12/14/2023] [Indexed: 12/19/2023]
Abstract
Senile osteoporosis increases fracture risks. Bone marrow stromal cells (BMSCs) are sensitive to aging. Deep insights into BMSCs aging are vital to elucidate the mechanisms underlying age-related bone loss. Recent advances showed that osteoporosis is associated with aberrant DNA methylation of many susceptible genes. Galectin-1 (Gal-1) has been proposed as a mediator of BMSCs functions. In our previous study, we showed that Gal-1 was downregulated in aged BMSCs and global deletion of Gal-1 in mice caused bone loss via impaired osteogenesis potential of BMSCs. Gal-1 promoter is featured by CpG islands. However, there are no reports concerning the DNA methylation status in Gal-1 promoter during osteoporosis. In the current study, we sought to investigate the role of DNA methylation in Gal-1 downregulation in aged BMSCs. The potential for anti-bone loss therapy based on modulating DNA methylation is explored. Our results showed that Dnmt3b-mediated Gal-1 promoter DNA hypermethylation plays an important role in Gal-1 downregulation in aged BMSCs, which inhibited β-catenin binding on Gal-1 promoter. Bone loss of aged mice was alleviated in response to in vivo deletion of Dnmt3b from BMSCs. Finally, when bone marrow of young wild-type (WT) mice or young Dnmt3bPrx1-Cre mice was transplanted into aged WT mice, Gal-1 level in serum and trabecular bone mass were elevated in recipient aged WT mice. Our study will benefit for deeper insights into the regulation mechanisms of Gal-1 expression in BMSCs during osteoporosis development, and for the discovery of new therapeutic targets for osteoporosis via modulating DNA methylation status.NEW & NOTEWORTHY There is Dnmt3b-mediated DNA methylation in Gal-1 promoter in aged bone marrow stromal cell (BMSC). DNA methylation causes Gal-1 downregulation and osteogenesis attenuation of aged BMSC. DNA methylation blocks β-catenin binding on Gal-1 promoter. Bone loss of aged mice is alleviated by in vivo deletion of Dnmt3b from BMSC.
Collapse
Affiliation(s)
- Liang Tang
- Department of Orthopedic Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yang-Yang Zhang
- Department of Orthopedic Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Wen-Jun Liu
- Department of Orthopedic Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Qiang Fu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jian Zhao
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China
| | - Yan-Bin Liu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
5
|
Szybiak W, Kujawa B, Miedziaszczyk M, Lacka K. Effect of Growth Hormone and Estrogen Replacement Therapy on Bone Mineral Density in Women with Turner Syndrome: A Meta-Analysis and Systematic Review. Pharmaceuticals (Basel) 2023; 16:1320. [PMID: 37765128 PMCID: PMC10536543 DOI: 10.3390/ph16091320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/27/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Osteoporosis is a serious implication of Turner syndrome (TS). Common methods for the treatment of TS are growth hormone (GHT) and estrogen replacement therapy (ERT). We examined the relationship between the treatment of TS and bone mineral density (BMD) of the lumbar spine. The purpose of our study was to show the currency of BMD states among patients with TS for treatment with GHT and ERT. We searched databases for studies published from inception to April 2023. The articles were related to TS, osteoporosis, ERT, GHT, BMD and treatment patients with TS. We applied the selection criteria: lumbar spine values at L1-L4; dual-energy X-ray absorptiometry (DXA); treatment which was applied: one group of articles: ERT and two group of articles: GHT; results performed as means ± SD. In total, 79 articles were analyzed, of which 20 studies were included and 5 were considered for meta-analysis. The total number of women in the articles selected was 71. Based on the results of the meta-analysis, the effect of ERT on BMD demonstrated a significant increase in BMD (the standardized mean difference in the random model was 0.593 g/cm2, 95% CI: 0.0705 to 1.116; p = 0.026), which showed that treatment with estrogen particularly increases bone mass during treatment, which contributes to reducing the risk of fractures. The effect of GHT on BMD demonstrated a non-significant decrease in BMD in patients with TS. The results for growth hormone show that this therapy does not improve bone density. However, our review emphasizes the beneficial effect of supplementing growth hormone (GH) on the clinical presentation of TS.
Collapse
Affiliation(s)
- Weronika Szybiak
- Students’ Scientific Section at the Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Science, 60-355 Poznan, Poland; (W.S.); (B.K.)
| | - Barbara Kujawa
- Students’ Scientific Section at the Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Science, 60-355 Poznan, Poland; (W.S.); (B.K.)
| | - Miłosz Miedziaszczyk
- Department of Nephrology, Transplantology and Internal Medicine, Poznan University of Medical Science, 60-355 Poznan, Poland;
| | - Katarzyna Lacka
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Science, 60-355 Poznan, Poland
| |
Collapse
|
6
|
Aguilar A, Gifre L, Ureña-Torres P, Carrillo-López N, Rodriguez-García M, Massó E, da Silva I, López-Báez V, Sánchez-Bayá M, Prior-Español Á, Urrutia M, Paul J, Bustos MC, Vila A, Garnica-León I, Navarro-González JF, Mateo L, Bover J. Pathophysiology of bone disease in chronic kidney disease: from basics to renal osteodystrophy and osteoporosis. Front Physiol 2023; 14:1177829. [PMID: 37342799 PMCID: PMC10277623 DOI: 10.3389/fphys.2023.1177829] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Chronic kidney disease (CKD) is a highly prevalent disease that has become a public health problem. Progression of CKD is associated with serious complications, including the systemic CKD-mineral and bone disorder (CKD-MBD). Laboratory, bone and vascular abnormalities define this condition, and all have been independently related to cardiovascular disease and high mortality rates. The "old" cross-talk between kidney and bone (classically known as "renal osteodystrophies") has been recently expanded to the cardiovascular system, emphasizing the importance of the bone component of CKD-MBD. Moreover, a recently recognized higher susceptibility of patients with CKD to falls and bone fractures led to important paradigm changes in the new CKD-MBD guidelines. Evaluation of bone mineral density and the diagnosis of "osteoporosis" emerges in nephrology as a new possibility "if results will impact clinical decisions". Obviously, it is still reasonable to perform a bone biopsy if knowledge of the type of renal osteodystrophy will be clinically useful (low versus high turnover-bone disease). However, it is now considered that the inability to perform a bone biopsy may not justify withholding antiresorptive therapies to patients with high risk of fracture. This view adds to the effects of parathyroid hormone in CKD patients and the classical treatment of secondary hyperparathyroidism. The availability of new antiosteoporotic treatments bring the opportunity to come back to the basics, and the knowledge of new pathophysiological pathways [OPG/RANKL (LGR4); Wnt-ß-catenin pathway], also affected in CKD, offers great opportunities to further unravel the complex physiopathology of CKD-MBD and to improve outcomes.
Collapse
Affiliation(s)
- Armando Aguilar
- Autonomous University of Chiapas, Tuxtla Gutiérrez, Mexico
- Department of Nephrology, Mexican Social Security, IMSS General Hospital of Zone No 2, Tuxtla Gutiérrez, Mexico
| | - Laia Gifre
- Department of Rheumatology, Hospital Germans Trias i Pujol, Badalona (Barcelona), Catalonia, Spain
| | - Pablo Ureña-Torres
- AURA Saint Ouen, Department of Nephrology and Dialysis and Department of Renal Physiology, Necker Hospital, University of Paris Descartes, Paris, France
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Minerva Rodriguez-García
- Nephrology Clinical Management Unit, Central University Hospital of Asturias (HUCA), Oviedo, Asturias, Spain
| | - Elisabeth Massó
- Department of Nephrology, University Hospital Germans Trias i Pujol (HGiTP), Badalona (Barcelona), Catalonia, Spain
- REMAR-IGTP Group, Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona (Barcelona), Catalonia, Spain
| | - Iara da Silva
- Department of Nephrology, University Hospital Germans Trias i Pujol (HGiTP), Badalona (Barcelona), Catalonia, Spain
- REMAR-IGTP Group, Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona (Barcelona), Catalonia, Spain
| | - Víctor López-Báez
- Department of Nephrology, University Hospital Germans Trias i Pujol (HGiTP), Badalona (Barcelona), Catalonia, Spain
- REMAR-IGTP Group, Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona (Barcelona), Catalonia, Spain
| | - Maya Sánchez-Bayá
- Department of Nephrology, University Hospital Germans Trias i Pujol (HGiTP), Badalona (Barcelona), Catalonia, Spain
- REMAR-IGTP Group, Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona (Barcelona), Catalonia, Spain
| | - Águeda Prior-Español
- Department of Rheumatology, Hospital Germans Trias i Pujol, Badalona (Barcelona), Catalonia, Spain
| | - Marina Urrutia
- Department of Nephrology, University Hospital Germans Trias i Pujol (HGiTP), Badalona (Barcelona), Catalonia, Spain
- REMAR-IGTP Group, Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona (Barcelona), Catalonia, Spain
| | - Javier Paul
- Department of Nephrology, University Hospital Germans Trias i Pujol (HGiTP), Badalona (Barcelona), Catalonia, Spain
- REMAR-IGTP Group, Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona (Barcelona), Catalonia, Spain
| | - Misael C. Bustos
- Department of Nephrology, Pontificia Catholic University of Chile, Santiago, Chile
| | - Anna Vila
- Department of Nephrology, University Hospital Germans Trias i Pujol (HGiTP), Badalona (Barcelona), Catalonia, Spain
- REMAR-IGTP Group, Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona (Barcelona), Catalonia, Spain
| | - Isa Garnica-León
- Department of Nephrology, Mexican Social Security, IMSS General Hospital of Zone No 2, Tuxtla Gutiérrez, Mexico
| | - Juan F. Navarro-González
- Research Unit and Nephrology Service, University Hospital of Nuestra Señora de la Candelaria, Santa Cruz de Tenerife, Islas Canarias, Spain
- Instituto de Tecnologías Biomédicas, Universidad de la Laguna, Islas Canarias, Spain
| | - Lourdes Mateo
- Department of Rheumatology, Hospital Germans Trias i Pujol, Badalona (Barcelona), Catalonia, Spain
| | - Jordi Bover
- Department of Nephrology, University Hospital Germans Trias i Pujol (HGiTP), Badalona (Barcelona), Catalonia, Spain
- REMAR-IGTP Group, Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona (Barcelona), Catalonia, Spain
| |
Collapse
|
7
|
Alekos NS, Kushwaha P, Kim SP, Li Z, Abood A, Dirckx N, Aja S, Kodama J, Garcia-Diaz JG, Otsuru S, Rendina-Ruedy E, Wolfgang MJ, Riddle RC. Mitochondrial β-oxidation of adipose-derived fatty acids by osteoblasts fuels parathyroid hormone-induced bone formation. JCI Insight 2023; 8:e165604. [PMID: 36729662 PMCID: PMC10070112 DOI: 10.1172/jci.insight.165604] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/01/2023] [Indexed: 02/03/2023] Open
Abstract
The energetic costs of bone formation require osteoblasts to coordinate their activities with tissues, like adipose, that can supply energy-dense macronutrients. In the case of intermittent parathyroid hormone (PTH) treatment, a strategy used to reduce fracture risk, bone formation is preceded by a change in systemic lipid homeostasis. To investigate the requirement for fatty acid oxidation by osteoblasts during PTH-induced bone formation, we subjected mice with osteoblast-specific deficiency of mitochondrial long-chain β-oxidation as well as mice with adipocyte-specific deficiency for the PTH receptor or adipose triglyceride lipase to an anabolic treatment regimen. PTH increased the release of fatty acids from adipocytes and β-oxidation by osteoblasts, while the genetic mouse models were resistant to the hormone's anabolic effect. Collectively, these data suggest that PTH's anabolic actions require coordinated signaling between bone and adipose, wherein a lipolytic response liberates fatty acids that are oxidized by osteoblasts to fuel bone formation.
Collapse
Affiliation(s)
- Nathalie S. Alekos
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Priyanka Kushwaha
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Soohyun P. Kim
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhu Li
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Abdullah Abood
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Naomi Dirckx
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joe Kodama
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jean G. Garcia-Diaz
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Satoru Otsuru
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth Rendina-Ruedy
- Department of Medicine and Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael J. Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ryan C. Riddle
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research & Development Service, Baltimore Veterans Administration Medical Center, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Yu D, Zhang S, Ma C, Huang S, Xu L, Liang J, Li H, Fan Q, Liu G, Zhai Z. CCL3 in the bone marrow microenvironment causes bone loss and bone marrow adiposity in aged mice. JCI Insight 2023; 8:159107. [PMID: 36378535 PMCID: PMC9870077 DOI: 10.1172/jci.insight.159107] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
The central physiological role of the bone marrow renders bone marrow stromal cells (BMSCs) particularly sensitive to aging. With bone aging, BMSCs acquire a differentiation potential bias in favor of adipogenesis over osteogenesis, and the underlying molecular mechanisms remain unclear. Herein, we investigated the factors underlying age-related changes in the bone marrow and their roles in BMSCs' differentiation. Antibody array revealed that CC chemokine ligand 3 (CCL3) accumulation occurred in the serum of naturally aged mice along with bone aging phenotypes, including bone loss, bone marrow adiposity, and imbalanced BMSC differentiation. In vivo Ccl3 deletion could rescue these phenotypes in aged mice. CCL3 improved the adipogenic differentiation potential of BMSCs, with a positive feedback loop between CCL3 and C/EBPα. CCL3 activated C/EBPα expression via STAT3, while C/EBPα activated CCL3 expression through direct promoter binding, facilitated by DNA hypomethylation. Moreover, CCL3 inhibited BMSCs' osteogenic differentiation potential by blocking β-catenin activity mediated by ERK-activated Dickkopf-related protein 1 upregulation. Blocking CCL3 in vivo via neutralizing antibodies ameliorated trabecular bone loss and bone marrow adiposity in aged mice. This study provides insights regarding age-related bone loss and bone marrow adiposity pathogenesis and lays a foundation for the identification of new targets for senile osteoporosis treatment.
Collapse
Affiliation(s)
- Degang Yu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuhong Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Ma
- Department of Orthopedics, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University; Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, The Xuzhou School of Clinical Medicine of Nanjing Medical University; and Xuzhou Central Hospital Affiliated to Medical School of Southeast University, Xuzhou, China
| | - Sen Huang
- Department of Orthopedics, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University; Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, The Xuzhou School of Clinical Medicine of Nanjing Medical University; and Xuzhou Central Hospital Affiliated to Medical School of Southeast University, Xuzhou, China
| | - Long Xu
- Department of Orthopedics, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University; Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, The Xuzhou School of Clinical Medicine of Nanjing Medical University; and Xuzhou Central Hospital Affiliated to Medical School of Southeast University, Xuzhou, China
| | - Jun Liang
- Department of Orthopedics, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University; Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, The Xuzhou School of Clinical Medicine of Nanjing Medical University; and Xuzhou Central Hospital Affiliated to Medical School of Southeast University, Xuzhou, China
| | - Huiwu Li
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiming Fan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guangwang Liu
- Department of Orthopedics, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University; Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, The Xuzhou School of Clinical Medicine of Nanjing Medical University; and Xuzhou Central Hospital Affiliated to Medical School of Southeast University, Xuzhou, China
| | - Zanjing Zhai
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Bandeira L, Lewiecki EM. Anabolic therapy for osteoporosis: update on efficacy and safety. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2022; 66:707-716. [PMID: 36382760 PMCID: PMC10118815 DOI: 10.20945/2359-3997000000566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Anabolic agents for the treatment of osteoporosis increase bone density, improve bone strength, and reduce fracture risk. They are distinguished from antiresorptive drugs by their property of increasing osteoblastic bone formation. Teriparatide and abaloparatide are parathyroid hormone receptor agonists that increase bone remodeling with bone formation increasing more than bone resorption. Romosozumab is a humanized monoclonal antibody to sclerostin that has a "dual effect" of increasing bone formation while decreasing bone resorption. The bone forming effects of anabolic therapy appear to be self-limited, making it imperative that it be followed by antiresorptive therapy to enhance or consolidate the beneficial effects achieved. Teriparatide, abaloparatide, and romosozumab each have unique pharmacological properties that must be appreciated when using them to treat patients at high risk for fracture. Clinical trials have shown a favorable balance of expected benefits and possible risks. Anabolic therapy is superior to bisphosphonates for high-risk patients, with greater benefit when initial treatment is with an anabolic agent followed by an antiresorptive drug, rather than the reverse sequence of therapy. Recent clinical practice guidelines have included recommendations with examples of patients who are candidates with anabolic therapy.
Collapse
|
10
|
Raimondi V, Toscani D, Marchica V, Burroughs-Garcia J, Storti P, Giuliani N. Metabolic features of myeloma cells in the context of bone microenvironment: Implication for the pathophysiology and clinic of myeloma bone disease. Front Oncol 2022; 12:1015402. [PMID: 36313705 PMCID: PMC9608343 DOI: 10.3389/fonc.2022.1015402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by the accumulation of malignant plasma cells (PCs) into the bone marrow (BM). The complex interaction between the BM microenvironment and MM PCs can lead to severe impairment of bone remodeling. Indeed, the BM microenvironment exerts a critical role in the survival of malignant PCs. Growing evidence indicates that MM cells have several metabolic features including enhanced glycolysis and an increase in lactate production through the upregulation of glucose transporters and enzymes. More recently, it has been reported that MM cells arehighly glutamine addicted. Interestingly, these metabolic changes in MM cells may affect BM microenvironment cells by altering the differentiation process of osteoblasts from mesenchymal stromal cells. The identification of glutamine metabolism alterations in MM cells and bone microenvironment may provide a rationale to design new therapeutic approaches and diagnostic tools. The osteolytic lesions are the most frequent clinical features in MM patients, often characterized by pathological fractures and acute pain. The use of the newer imaging techniques such as Magnetic Resonance Imaging (MRI) and combined Positron Emission Tomography (PET) and Computerized Tomography (CT) has been introduced into clinical practice to better define the skeletal involvement. Currently, the PET/CT with 18F-fluorodeoxyglucose (FDG) is the diagnostic gold standard to detect active MM bone disease due to the high glycolytic activity of MM cells. However, new tracers are actively under investigation because a portion of MM patients remains negative at the skeletal level by 18F-FDG. In this review, we will summarize the existing knowledge on the metabolic alterations of MM cells considering their impact on the BM microenvironment cells and particularly in the subsequent formation of osteolytic bone lesions. Based on this, we will discuss the identification of possible new druggable targets and the use of novel metabolic targets for PET imaging in the detection of skeletal lesions, in the staging and treatment response of MM patients.
Collapse
Affiliation(s)
- Vincenzo Raimondi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | | | - Paola Storti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- *Correspondence: Paola Storti, ; Nicola Giuliani,
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Hematology, “Azienda Ospedaliero-Universitaria di Parma”, Parma, Italy
- *Correspondence: Paola Storti, ; Nicola Giuliani,
| |
Collapse
|
11
|
Arthur A, Gronthos S. Eph-Ephrin Signaling Mediates Cross-Talk Within the Bone Microenvironment. Front Cell Dev Biol 2021; 9:598612. [PMID: 33634116 PMCID: PMC7902060 DOI: 10.3389/fcell.2021.598612] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/15/2021] [Indexed: 12/18/2022] Open
Abstract
Skeletal integrity is maintained through the tightly regulated bone remodeling process that occurs continuously throughout postnatal life to replace old bone and to repair skeletal damage. This is maintained primarily through complex interactions between bone resorbing osteoclasts and bone forming osteoblasts. Other elements within the bone microenvironment, including stromal, osteogenic, hematopoietic, endothelial and neural cells, also contribute to maintaining skeletal integrity. Disruption of the dynamic interactions between these diverse cellular systems can lead to poor bone health and an increased susceptibility to skeletal diseases including osteopenia, osteoporosis, osteoarthritis, osteomalacia, and major fractures. Recent reports have implicated a direct role for the Eph tyrosine kinase receptors and their ephrin ligands during bone development, homeostasis and skeletal repair. These membrane-bound molecules mediate contact-dependent signaling through both the Eph receptors, termed forward signaling, and through the ephrin ligands, referred to as reverse signaling. This review will focus on Eph/ ephrin cross-talk as mediators of hematopoietic and stromal cell communication, and how these interactions contribute to blood/ bone marrow function and skeletal integrity during normal steady state or pathological conditions.
Collapse
Affiliation(s)
- Agnieszka Arthur
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
12
|
Estell EG, Rosen CJ. Emerging insights into the comparative effectiveness of anabolic therapies for osteoporosis. Nat Rev Endocrinol 2021; 17:31-46. [PMID: 33149262 DOI: 10.1038/s41574-020-00426-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2020] [Indexed: 01/01/2023]
Abstract
Over the past three decades, the mainstay of treatment for osteoporosis has been antiresorptive agents (such as bisphosphonates), which have been effective with continued administration in lowering fracture risk. However, the clinical landscape has changed as adherence to these medications has declined due to perceived adverse effects. As a result, decreases in hip fracture rates that followed the introduction of bisphosphonates have now levelled off, which is coincident with a decline in the use of the antiresorptive agents. In the past two decades, two types of anabolic agents (including three new drugs), which represent a novel approach to improving bone quality by increasing bone formation, have been approved. These therapies are expected to lead to a new clinical paradigm in which anabolic agents will be used either alone or in combination with antiresorptive agents to build new bone and reduce fracture risk. This Review examines the mechanisms of action for these anabolic agents by detailing their receptor-activating properties for key cell types in the bone and marrow niches. Using these advances in bone biology as context, the comparative effectiveness of these anabolic agents is discussed in relation to other therapeutic options for osteoporosis to better guide their clinical application in the future.
Collapse
Affiliation(s)
- Eben G Estell
- Maine Medical Center Research Institute, Scarborough, ME, USA
| | | |
Collapse
|
13
|
Li JY, Yu M, Pal S, Tyagi AM, Dar H, Adams J, Weitzmann MN, Jones RM, Pacifici R. Parathyroid hormone-dependent bone formation requires butyrate production by intestinal microbiota. J Clin Invest 2020; 130:1767-1781. [PMID: 31917685 DOI: 10.1172/jci133473] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/23/2019] [Indexed: 12/22/2022] Open
Abstract
Parathyroid hormone (PTH) is a critical regulator of skeletal development that promotes both bone formation and bone resorption. Using microbiota depletion by wide-spectrum antibiotics and germ-free (GF) female mice, we showed that the microbiota was required for PTH to stimulate bone formation and increase bone mass. Microbiota depletion lowered butyrate levels, a metabolite responsible for gut-bone communication, while reestablishment of physiologic levels of butyrate restored PTH-induced anabolism. The permissive activity of butyrate was mediated by GPR43 signaling in dendritic cells and by GPR43-independent signaling in T cells. Butyrate was required for PTH to increase the number of bone marrow (BM) regulatory T cells (Tregs). Tregs stimulated production of the osteogenic Wnt ligand Wnt10b by BM CD8+ T cells, which activated Wnt-dependent bone formation. Together, these data highlight the role that butyrate produced by gut luminal microbiota plays in triggering regulatory pathways, which are critical for the anabolic action of PTH in bone.
Collapse
Affiliation(s)
- Jau-Yi Li
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and.,Emory Microbiome Center, Emory University, Atlanta, Georgia, USA
| | - Mingcan Yu
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and.,Emory Microbiome Center, Emory University, Atlanta, Georgia, USA
| | - Subhashis Pal
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and.,Emory Microbiome Center, Emory University, Atlanta, Georgia, USA
| | - Abdul Malik Tyagi
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and.,Emory Microbiome Center, Emory University, Atlanta, Georgia, USA
| | - Hamid Dar
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and.,Emory Microbiome Center, Emory University, Atlanta, Georgia, USA
| | - Jonathan Adams
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and.,Emory Microbiome Center, Emory University, Atlanta, Georgia, USA
| | - M Neale Weitzmann
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and.,Emory Microbiome Center, Emory University, Atlanta, Georgia, USA.,Atlanta Department of Veterans Affairs Medical Center, Decatur, Georgia, USA
| | - Rheinallt M Jones
- Emory Microbiome Center, Emory University, Atlanta, Georgia, USA.,Division of Pediatric Gastroenterology,, Hepatology, and Nutrition, Department of Pediatrics, and.,Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, Georgia, USA
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and.,Emory Microbiome Center, Emory University, Atlanta, Georgia, USA.,Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Kosugi K, Tajima T, Menuki K, Okuma KF, Tokuda K, Fukuda H, Okada Y, Tsukamoto M, Yamanaka Y, Zenke Y, Sakai A. Disruption of the aldehyde dehydrogenase 2 gene increases the bone anabolic response to intermittent PTH treatment in an ovariectomized mouse model. Bone 2020; 136:115370. [PMID: 32325250 DOI: 10.1016/j.bone.2020.115370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 12/14/2022]
Abstract
Aldehyde dehydrogenase 2 (ALDH2) is the enzyme that oxidizes the acetaldehyde produced by alcohol metabolism. This variant not only affects the response to alcohol but is also associated with several diseases, such as esophageal cancer, myocardial infarction, and particularly osteoporosis. In our previous study, we reported that compared to wild-type (WT) mice, Aldh2 knockout (KO) mice naturally have a strong bone formation ability, and high expression of parathyroid hormone receptor (PTHR1) in osteocytes. The effect of the Aldh2 gene on bone metabolism in response to intermittent PTH treatment is unknown. The purpose of this study was to clarify the effect of the Aldh2 gene on the bone anabolic response to intermittent PTH treatment in ovariectomized mice. Female KO and WT mice were ovariectomized at 8 weeks of age. At 14 weeks of age, the KO and WT mice were divided into vehicle-treated (Veh) and PTH-treated (PTH) groups (i.e., the WT-Veh, WT-PTH, KO-Veh and KO-PTH groups). PTH (1-34) and vehicle were subcutaneously administered to each group at a dose of 40 μg/kg body weight (BW) five times per week for 4 weeks. Micro-CT showed that the bone volume (BV), trabecular number (Tb.N), connectivity density (Conn.D), and cortical thickness (Ct.Th) values in the KO-PTH mice were significantly higher than those in the KO-Veh mice. Histomorphometric analysis showed that the BV, Tb.N, and mineral apposition rate (MAR) values in the KO-PTH group were significantly higher than those in the KO-Veh group. The mRNA expression level of PTHR1 in the KO-PTH group was significantly increased and that of p21 in the KO-PTH group was significantly decreased compared with the levels in the KO-Veh group. The expression of PTHR in osteocytes from the KO-PTH group was also significantly increased compared with that in osteocytes from the KO-Veh group. Furthermore, cell cultures revealed that the ALP+CFU-f/total CFU-f percentage was significantly higher in the KO-PTH group than in the KO-Veh group. We concluded that in ovariectomized Aldh2 KO mice, the bone anabolic response to intermittent PTH treatment was significantly enhanced compared to that in WT mice, which may be mediated by the high expression level of PTHR1.
Collapse
Affiliation(s)
- Kenji Kosugi
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Takafumi Tajima
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan.
| | - Kunitaka Menuki
- Department of Orthopaedic Surgery, Kitakyushu Municipal Yahata Hospital, 2-6-2 Ogura, Yahatahigashi-ku, Kitakyushu 805-8534, Japan
| | - Kayoko Furukawa Okuma
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Kotaro Tokuda
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Hokuto Fukuda
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Yasuaki Okada
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Manabu Tsukamoto
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Yoshiaki Yamanaka
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Yukichi Zenke
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Akinori Sakai
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| |
Collapse
|
15
|
Abstract
Post-natal bone development is characterized by substantial longitudinal bone growth and changes in skeletal size and shape. Bone is in a dynamic process of continuous remodeling which helps to regulate calcium homeostasis, repair micro-damage to bones from everyday stress, and to shape the skeleton during growth. Bone growth is regulated by systemic hormones and locally generated factors. Understanding their mechanisms of action enables us to obtain a better appreciation of the cellular and molecular basis of bone remodeling and could therefore be valuable in approaches to new therapies. This article will review molecular and cellular control of skeletal growth in the post-natal period, the physiology of each bone cell with their systemic and local regulators, as well as the physiology of bone remodeling.
Collapse
Affiliation(s)
- Rania Ali El-Farrash
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Abbassya Square, 11566, Cairo, Egypt.
| | - Radwa Hassan Ali
- Physiology Department, Faculty of Medicine, Ain Shams University, Abbassya Square, 11566, Cairo, Egypt.
| | - Noha Mokhtar Barakat
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Abbassya Square, 11566, Cairo, Egypt.
| |
Collapse
|
16
|
Abstract
Osteoblasts are specialized mesenchymal cells that synthesize bone matrix and coordinate the mineralization of the skeleton. These cells work in harmony with osteoclasts, which resorb bone, in a continuous cycle that occurs throughout life. The unique function of osteoblasts requires substantial amounts of energy production, particularly during states of new bone formation and remodelling. Over the last 15 years, studies have shown that osteoblasts secrete endocrine factors that integrate the metabolic requirements of bone formation with global energy balance through the regulation of insulin production, feeding behaviour and adipose tissue metabolism. In this article, we summarize the current understanding of three osteoblast-derived metabolic hormones (osteocalcin, lipocalin and sclerostin) and the clinical evidence that suggests the relevance of these pathways in humans, while also discussing the necessity of specific energy substrates (glucose, fatty acids and amino acids) to fuel bone formation and promote osteoblast differentiation.
Collapse
Affiliation(s)
- Naomi Dirckx
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Megan C Moorer
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Thomas L Clemens
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA.
| |
Collapse
|
17
|
Weng SJ, Xie ZJ, Wu ZY, Yan DY, Tang JH, Shen ZJ, Li H, Bai BL, Boodhun V, Eric Dong XD, Yang L. Effects of combined menaquinone-4 and PTH 1-34 treatment on osetogenesis and angiogenesis in calvarial defect in osteopenic rats. Endocrine 2019; 63:376-384. [PMID: 30244350 DOI: 10.1007/s12020-018-1761-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 09/11/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE The aim of this study was to evaluate the effect of combining human parathyroid hormone (1-34) (PTH1-34; PTH) and menaquinone-4 (MK-4) on calvarial bone defect repair in osteopenic rats. METHODS Fourteen week olds were subject to craniotomy for the establishment of osteopenic animal models fed through a chronically low-protein diet. After that, critical calvarial defect model was established and all rats were randomly divided into four groups: sham, MK-4, PTH, and PTH + MK-4. The animals received MK-4 (30 mg/kg/day), PTH1-34 (60 μg/kg, three times a week), or PTH1-34 (60 μg/kg, three times a week) plus MK-4 (30 mg/kg/day) for 8 weeks, respectively. Serum γ-carboxylated osteocalcin (Gla-OC) levels, histological and immunofluorescent labeling were employed to evaluate the bone formation and mineralization in calvarial bone defect. In addition, Microfil perfusion, immunohistochemical, and micro-CT suggested enhanced angiogenesis and bone formation in calvarial bone healing. RESULTS In this study, treatment with either PTH1-34 or MK-4 promoted bone formation and vascular formation in calvarial bone defects compared with the sham group. In addition, combined treatment of PTH1-34 plus MK-4 increased serum level of Gla-OC, improved vascular number and vascular density, and enhanced bone formation in calvarial bone defect in osteopenic conditions as compared with monotherapy. CONCLUSIONS In summary, this study indicated that PTH1-34 plus MK-4 combination therapy accelerated bone formation and angiogenesis in calvarial bone defects in presence of osteopenia.
Collapse
MESH Headings
- Animals
- Bone Diseases, Metabolic/complications
- Bone Diseases, Metabolic/diagnosis
- Bone Diseases, Metabolic/drug therapy
- Bone Diseases, Metabolic/pathology
- Drug Therapy, Combination
- Female
- Fracture Healing/drug effects
- Fractures, Spontaneous/diagnosis
- Fractures, Spontaneous/drug therapy
- Fractures, Spontaneous/etiology
- Fractures, Spontaneous/pathology
- Neovascularization, Physiologic/drug effects
- Osteogenesis/drug effects
- Parathyroid Hormone/administration & dosage
- Rats
- Rats, Sprague-Dawley
- Skull/diagnostic imaging
- Skull/drug effects
- Skull/injuries
- Skull/pathology
- Skull Fractures/diagnosis
- Skull Fractures/drug therapy
- Skull Fractures/etiology
- Skull Fractures/pathology
- Vitamin K 2/administration & dosage
- Vitamin K 2/analogs & derivatives
- X-Ray Microtomography
Collapse
Affiliation(s)
- She-Ji Weng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhong-Jie Xie
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zong-Yi Wu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - De-Yi Yan
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia-Hao Tang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zi-Jian Shen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hang Li
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bing-Li Bai
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Viraj Boodhun
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiang Da Eric Dong
- Department of Surgery, Westchester Medical Center / New York Medical College, Valhalla, NY, USA
| | - Lei Yang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
18
|
Liu H, Su H, Wang X, Hao W. MiR-148a regulates bone marrow mesenchymal stem cells-mediated fracture healing by targeting insulin-like growth factor 1. J Cell Biochem 2019; 120:1350-1361. [PMID: 30335895 DOI: 10.1002/jcb.27121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/04/2018] [Indexed: 01/24/2023]
Abstract
The purpose of this study was to investigate the underlying molecular mechanisms of fracture healing mediated by bone marrow mesenchymal stem cells. Differentially expressed microRNAs in acutely injured subjects and healthy volunteers were screened by microarray analysis. The dual luciferase reporter system was used to verify whether insulin-like growth factor 1 (IGF1) was the direct target gene regulated by miR-148a. The expression level of miR-148a and IGF1 after osteogenic differentiation was detected by quantitative real-time polymerase chain reaction. Western blot was used to determine the protein expression of bone markers, including IGF1, runt-related transcription factor 2 (Runx2), osteocalcin, and osteopontin in rat bone marrow-derived mesenchymal stem cells. Alkaline phosphatase and alizarin red staining was used to detect alkaline phosphatase activity and calcium deposition. An animal fracture model was used for in vivo experiments. MiR-148a was highly expressed in acutely injured subjects compared with healthy volunteers, and IGF1 was a target of miR-148a. Moreover, compared with the negative control group, IGF1 messenger RNA expression was significantly increased in the miR-148a antagomir group. During osteogenic differentiation, the expression of IGF1, Runx2, osteocalcin, and osteopontin was higher in the miR-148a antagomir group than other groups. In vivo experiments further confirmed that upregulation of IGF1 enhanced fracture healing efficiently by decreasing callus width and area and improving bone mineral density, maximum load, stiffness, and energy absorption. It was proved that IGF1 was the direct target gene of miR-148a, and the use of rat bone marrow-derived mesenchymal stem cells with low expression of miR-148a could improve fracture healing by upregulating IGF1.
Collapse
Affiliation(s)
- Hongzhi Liu
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Hao Su
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Xin Wang
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Wei Hao
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| |
Collapse
|
19
|
Combined treatment with vitamin K2 and PTH enhanced bone formation in ovariectomized rats and increased differentiation of osteoblast in vitro. Chem Biol Interact 2019; 300:101-110. [PMID: 30639440 DOI: 10.1016/j.cbi.2019.01.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 12/22/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
Osteoporosis is accompanied by insufficient osteogenic capacity. Several lines of evidence suggested that solutions to enhance osteoblastogenesis were important strategies for osteoporotic bone defect repair. This study investigated the effect of combined treatment with vitamin K2 and PTH on bone formation in calvarial bone defect in osteoporotic rats and its influence on osteoblast in vitro. Bilateral ovariectomy was used in SPF Sprague Dawley rats to generate an osteoporosis model. Subsequently, a calvarial defect model was established and all osteoporotic rats were randomly assigned to the following groups: control, VK (vitamin K2, 30 mg/kg everyday), PTH (recombinant human PTH (1-34), 60 μg/kg, three times a week) or VK + PTH (vitamin K2, 30 mg/kg everyday plus PTH, 60 μg/kg three times a week) for 8 weeks. In vitro, bone marrow-derived stem cells (BMSCs) were cultured and treated with vitamin K2, PTH or vitamin K2+PTH. ALP staining and western blot were performed to observe the influence of combined treatment on BMSCs. Bone formation within calvarial defect were assessed by serum γ-carboxylated osteocalcin (Gla-OC), micro-CT, histological and immunofluorescent labeling. In this study, combined treatment of PTH and vitamin K2 showed positive effects on preventing bone loss in femurs in OVX rats. Combined treatment increased serum Gla-OC and promoted bone formation in osteoporotic calvarial bone defects. Immunohistochemistry showed that OCN and RUNX2 were more highly expressed in the VK + PTH group than in the control groups. In vitro studies results suggested that combined treatment with PTH and vitamin K2 increased expression of ALP, BMP2 and RUNX2 in BMSCs. Our data suggested that the combination of vitamin K2 and PTH increased differentiation of osteoblast and had a synergistic effect on bone formation in osteoporotic calvarial bone defect.
Collapse
|
20
|
Karvande A, Kushwaha P, Ahmad N, Adhikary S, Kothari P, Tripathi AK, Khedgikar V, Trivedi R. Glucose dependent miR-451a expression contributes to parathyroid hormone mediated osteoblast differentiation. Bone 2018; 117:98-115. [PMID: 30218791 DOI: 10.1016/j.bone.2018.09.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023]
Abstract
Parathyroid hormone (PTH; amino acid 1-34, known as teriparatide) has reported promoting differentiation and glucose uptake in osteoblasts. However, how PTH regulates glucose metabolism to facilitate osteoblast differentiation is not understood. Here, we report that PTH promotes glucose dependent miR-451a expression which stimulates osteoblast differentiation. In addition to glucose uptake, PTH suppresses AMPK phosphorylation via PI3K-mTOR-AKT axis thereby preventing phosphorylation and inactivation of octamer-binding transcription factor 1 (OCT-1) which has been reported to act on the promoter region of miR-451a. Modulation of AMPK activity controls miR-451a levels in differentiating osteoblasts. Moreover, pharmacological inhibition of PI3K-mTOR-AKT axis suppressed miR-451a via increased AMPK activity. We report that this glucose regulated miRNA is an anabolic target and transfection of miR-451a mimic induces osteoblast differentiation and mineralization in vitro. These actions were mediated through the suppression of Odd-skipped related 1 (Osr1) and activation of Runx2 transcription. When injected in vivo, the miR-451a mimic significantly increased osteoblastogenesis, mineralization, reversed ovariectomy induced bone loss and improved bone strength. Together, these findings suggest that enhanced osteoblast differentiation associated with bone formation in case of PTH therapy is also a consequence of elevated miR-451a levels via glucose regulation. Consequently, this miRNA has the potential to be a therapeutic target for conditions of bone loss.
Collapse
Affiliation(s)
- Anirudha Karvande
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Priyanka Kushwaha
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Naseer Ahmad
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Sulekha Adhikary
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Priyanka Kothari
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Ashish Kumar Tripathi
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Vikram Khedgikar
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Ritu Trivedi
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India.
| |
Collapse
|
21
|
Xu ZS, Dai F, Chen J, Lv M, Cheng JW, Zhang XM, Lin BW. Experimental research into the potential therapeutic effect of GYY4137 on Ovariectomy-induced osteoporosis. Cell Mol Biol Lett 2018; 23:47. [PMID: 30305826 PMCID: PMC6167887 DOI: 10.1186/s11658-018-0114-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 09/26/2018] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Evidence has shown that endogenous H2S plays an important role in the physiological and pathophysiological processes of many organs. The study aimed to explore whether exogenous H2S has a potential therapeutic effect on a rat ovariectomy-induced model of osteoporosis. METHODS The OVX osteoporosis model was established in female Sprague-Dawley rats by full bilateral ovariectomy. The rats were randomly divided into four groups, with the two experimental groups receiving an intraperitoneal injection of GYY4137 or sodium alendronate. The level of H2S in the plasma was determined and common laboratory indicators to diagnose osteoporosis, such as alkaline phosphatase (ALP) activity and the levels of osteocalcin (OCN), calcitonin, parathyroid hormone and leptin were measured. The bone mineral density (BMD) of the 4th and 5th lumbar vertebrae was measured using dual-energy X-ray absorptiometry. The maximum stress of femoral fracture was obtained through a three-point bending test of the femur. RESULTS The OVX osteoporosis model was successfully established. GYY4137 was injected to increase the level of H2S in the plasma in one group, designated OVX-GYY during the observation period (p < 0.05). At 12 weeks, the BMD value of the fourth lumbar vertebra in the OVX-GYY group had increased (p < 0.05). The BMD femur value in the OVX-vehicle group had decreased (p < 0.05). Bilateral ovariectomy leads to biochemical disorders related to bone metabolism and hormone levels in rat plasma (all p < 0.05). Ovariectomy also reduced blood calcium, blood phosphate and calcitonin, and increased parathyroid hormone and leptin. The opposite results were obtained for the groups with alendronate sodium or GYY4137 treatment (all p < 0.05). CONCLUSIONS Through the slow release of H2S, GYY4137 did an excellent job of simulating endogenous neuroendocrine gaseous signaling molecules. Exogenous H2S had a regulatory effect on osteoporosis in ovariectomized rats, showing potential value for the treatment of human postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Zhong-Shi Xu
- Department of Orthopedics, Second Clinical Medical College of Jinan University (Shenzhen People’s Hospital), Dongmen North Road 1017, Luohu District, Shenzhen, 518020 China
| | - Feng Dai
- Department of Radiology, Second Clinical Medical College of Jinan University (Shenzhen People’s Hospital), Shenzhen, 518020 China
| | - Ji Chen
- Department of Orthopedics, Second Clinical Medical College of Jinan University (Shenzhen People’s Hospital), Dongmen North Road 1017, Luohu District, Shenzhen, 518020 China
| | - Meng Lv
- Department of Orthopedics, Second Clinical Medical College of Jinan University (Shenzhen People’s Hospital), Dongmen North Road 1017, Luohu District, Shenzhen, 518020 China
| | - Ji-Wu Cheng
- Department of Orthopedics, Second Clinical Medical College of Jinan University (Shenzhen People’s Hospital), Dongmen North Road 1017, Luohu District, Shenzhen, 518020 China
| | - Xiao-Ming Zhang
- Department of Orthopedics, Second Clinical Medical College of Jinan University (Shenzhen People’s Hospital), Dongmen North Road 1017, Luohu District, Shenzhen, 518020 China
| | - Bo-Wen Lin
- Department of Orthopedics, Second Clinical Medical College of Jinan University (Shenzhen People’s Hospital), Dongmen North Road 1017, Luohu District, Shenzhen, 518020 China
| |
Collapse
|
22
|
Wein MN, Kronenberg HM. Regulation of Bone Remodeling by Parathyroid Hormone. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a031237. [PMID: 29358318 DOI: 10.1101/cshperspect.a031237] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Parathyroid hormone (PTH) exerts profound effects on skeletal homeostasis through multiple cellular and molecular mechanisms. Continuous hyperparathyroidism causes net loss of bone mass, despite accelerating bone formation by osteoblasts. Intermittent treatment with PTH analogs represents the only Food and Drug Administration (FDA)-approved bone anabolic osteoporosis treatment strategy. Functional PTH receptors are present on cells of the osteoblast lineage, ranging from early skeletal stem cells to matrix-embedded osteocytes. In addition, bone remodeling by osteoclasts liberates latent growth factors present within bone matrix. Here, we will provide an overview of the multiple cellular and molecular mechanisms through which PTH influences bone homeostasis. Notably, net skeletal effects of continuous versus intermittent can differ significantly. Where possible, we will highlight mechanisms through which continuous hyperparathyroidism leads to bone loss, and through which intermittent hyperparathyroidism boosts bone mass. Given the therapeutic usage of intermittent PTH (iPTH) treatment for osteoporosis, particular attention will be paid toward mechanisms underlying the bone anabolic effects of once daily PTH administration.
Collapse
Affiliation(s)
- Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| | - Henry M Kronenberg
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| |
Collapse
|
23
|
Lindsey RC, Rundle CH, Mohan S. Role of IGF1 and EFN-EPH signaling in skeletal metabolism. J Mol Endocrinol 2018; 61:T87-T102. [PMID: 29581239 PMCID: PMC5966337 DOI: 10.1530/jme-17-0284] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 03/26/2018] [Indexed: 01/11/2023]
Abstract
Insulin-like growth factor 1(IGF1) and ephrin ligand (EFN)-receptor (EPH) signaling are both crucial for bone cell function and skeletal development and maintenance. IGF1 signaling is the major mediator of growth hormone-induced bone growth, but a host of different signals and factors regulate IGF1 signaling at the systemic and local levels. Disruption of the Igf1 gene results in reduced peak bone mass in both experimental animal models and humans. Additionally, EFN-EPH signaling is a complex system which, particularly through cell-cell interactions, contributes to the development and differentiation of many bone cell types. Recent evidence has demonstrated several ways in which the IGF1 and EFN-EPH signaling pathways interact with and depend upon each other to regulate bone cell function. While much remains to be elucidated, the interaction between these two signaling pathways opens a vast array of new opportunities for investigation into the mechanisms of and potential therapies for skeletal conditions such as osteoporosis and fracture repair.
Collapse
Affiliation(s)
- Richard C Lindsey
- Musculoskeletal Disease CenterVA Loma Linda Healthcare System, Loma Linda, California, USA
- Division of BiochemistryDepartment of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
- Center for Health Disparities and Molecular MedicineDepartment of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Charles H Rundle
- Musculoskeletal Disease CenterVA Loma Linda Healthcare System, Loma Linda, California, USA
- Department of MedicineLoma Linda University, Loma Linda, California, USA
| | - Subburaman Mohan
- Musculoskeletal Disease CenterVA Loma Linda Healthcare System, Loma Linda, California, USA
- Division of BiochemistryDepartment of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
- Center for Health Disparities and Molecular MedicineDepartment of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
- Department of MedicineLoma Linda University, Loma Linda, California, USA
| |
Collapse
|
24
|
Yakar S, Werner H, Rosen CJ. Insulin-like growth factors: actions on the skeleton. J Mol Endocrinol 2018; 61:T115-T137. [PMID: 29626053 PMCID: PMC5966339 DOI: 10.1530/jme-17-0298] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 12/20/2022]
Abstract
The discovery of the growth hormone (GH)-mediated somatic factors (somatomedins), insulin-like growth factor (IGF)-I and -II, has elicited an enormous interest primarily among endocrinologists who study growth and metabolism. The advancement of molecular endocrinology over the past four decades enables investigators to re-examine and refine the established somatomedin hypothesis. Specifically, gene deletions, transgene overexpression or more recently, cell-specific gene-ablations, have enabled investigators to study the effects of the Igf1 and Igf2 genes in temporal and spatial manners. The GH/IGF axis, acting in an endocrine and autocrine/paracrine fashion, is the major axis controlling skeletal growth. Studies in rodents have clearly shown that IGFs regulate bone length of the appendicular skeleton evidenced by changes in chondrocytes of the proliferative and hypertrophic zones of the growth plate. IGFs affect radial bone growth and regulate cortical and trabecular bone properties via their effects on osteoblast, osteocyte and osteoclast function. Interactions of the IGFs with sex steroid hormones and the parathyroid hormone demonstrate the significance and complexity of the IGF axis in the skeleton. Finally, IGFs have been implicated in skeletal aging. Decreases in serum IGFs during aging have been correlated with reductions in bone mineral density and increased fracture risk. This review highlights many of the most relevant studies in the IGF research landscape, focusing in particular on IGFs effects on the skeleton.
Collapse
Affiliation(s)
- Shoshana Yakar
- David B. Kriser Dental Center, Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010-4086, USA
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Scarborough, Maine 04074, USA
| |
Collapse
|
25
|
The art of building bone: emerging role of chondrocyte-to-osteoblast transdifferentiation in endochondral ossification. Bone Res 2018. [PMID: 29928541 DOI: 10.1038/s41413‐018‐0021‐z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There is a worldwide epidemic of skeletal diseases causing not only a public health issue but also accounting for a sizable portion of healthcare expenditures. The vertebrate skeleton is known to be formed by mesenchymal cells condensing into tissue elements (patterning phase) followed by their differentiation into cartilage (chondrocytes) or bone (osteoblasts) cells within the condensations. During the growth and remodeling phase, bone is formed directly via intramembranous ossification or through a cartilage to bone conversion via endochondral ossification routes. The canonical pathway of the endochondral bone formation process involves apoptosis of hypertrophic chondrocytes followed by vascular invasion that brings in osteoclast precursors to remove cartilage and osteoblast precursors to form bone. However, there is now an emerging role for chondrocyte-to-osteoblast transdifferentiation in the endochondral ossification process. Although the concept of "transdifferentiation" per se is not recent, new data using a variety of techniques to follow the fate of chondrocytes in different bones during embryonic and post-natal growth as well as during fracture repair in adults have identified three different models for chondrocyte-to-osteoblast transdifferentiation (direct transdifferentiation, dedifferentiation to redifferentiation, and chondrocyte to osteogenic precursor). This review focuses on the emerging models of chondrocyte-to-osteoblast transdifferentiation and their implications for the treatment of skeletal diseases as well as the possible signaling pathways that contribute to chondrocyte-to-osteoblast transdifferentiation processes.
Collapse
|
26
|
Aghajanian P, Mohan S. The art of building bone: emerging role of chondrocyte-to-osteoblast transdifferentiation in endochondral ossification. Bone Res 2018; 6:19. [PMID: 29928541 PMCID: PMC6002476 DOI: 10.1038/s41413-018-0021-z] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 12/21/2022] Open
Abstract
There is a worldwide epidemic of skeletal diseases causing not only a public health issue but also accounting for a sizable portion of healthcare expenditures. The vertebrate skeleton is known to be formed by mesenchymal cells condensing into tissue elements (patterning phase) followed by their differentiation into cartilage (chondrocytes) or bone (osteoblasts) cells within the condensations. During the growth and remodeling phase, bone is formed directly via intramembranous ossification or through a cartilage to bone conversion via endochondral ossification routes. The canonical pathway of the endochondral bone formation process involves apoptosis of hypertrophic chondrocytes followed by vascular invasion that brings in osteoclast precursors to remove cartilage and osteoblast precursors to form bone. However, there is now an emerging role for chondrocyte-to-osteoblast transdifferentiation in the endochondral ossification process. Although the concept of "transdifferentiation" per se is not recent, new data using a variety of techniques to follow the fate of chondrocytes in different bones during embryonic and post-natal growth as well as during fracture repair in adults have identified three different models for chondrocyte-to-osteoblast transdifferentiation (direct transdifferentiation, dedifferentiation to redifferentiation, and chondrocyte to osteogenic precursor). This review focuses on the emerging models of chondrocyte-to-osteoblast transdifferentiation and their implications for the treatment of skeletal diseases as well as the possible signaling pathways that contribute to chondrocyte-to-osteoblast transdifferentiation processes.
Collapse
Affiliation(s)
- Patrick Aghajanian
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California USA
- Department of Medicine, Loma Linda University, Loma Linda, California USA
- Department of Orthopedics, Loma Linda University, Loma Linda, California USA
- Department of Biochemistry, Loma Linda University, Loma Linda, California USA
| |
Collapse
|
27
|
IGF-I induced phosphorylation of PTH receptor enhances osteoblast to osteocyte transition. Bone Res 2018; 6:5. [PMID: 29507819 PMCID: PMC5827661 DOI: 10.1038/s41413-017-0002-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/25/2017] [Accepted: 05/02/2017] [Indexed: 02/05/2023] Open
Abstract
Parathyroid hormone (PTH) regulates bone remodeling by activating PTH type 1 receptor (PTH1R) in osteoblasts/osteocytes. Insulin-like growth factor type 1 (IGF-1) stimulates mesenchymal stem cell differentiation to osteoblasts. However, little is known about the signaling mechanisms that regulates the osteoblast-to-osteocyte transition. Here we report that PTH and IGF-I synergistically enhance osteoblast-to-osteocyte differentiation. We identified that a specific tyrosine residue, Y494, on the cytoplasmic domain of PTH1R can be phosphorylated by insulin-like growth factor type I receptor (IGF1R) in vitro. Phosphorylated PTH1R localized to the barbed ends of actin filaments and increased actin polymerization during morphological change of osteoblasts into osteocytes. Disruption of the phosphorylation site reduced actin polymerization and dendrite length. Mouse models with conditional ablation of PTH1R in osteoblasts demonstrated a reduction in the number of osteoctyes and dendrites per osteocyte, with complete overlap of PTH1R with phosphorylated-PTH1R positioning in osteocyte dendrites in wild-type mice. Thus, our findings reveal a novel signaling mechanism that enhances osteoblast-to-osteocyte transition by direct phosphorylation of PTH1R by IGF1R. A key hormone and growth factor work together to help turn bone-forming cells into mature bone. Janet Crane and colleagues from Johns Hopkins University School of Medicine in Baltimore, Maryland, USA, tested the effects of parathyroid hormone (PTH) and insulin like-growth factor type 1 (IGF-1) signaling on the differentiation of bone-forming osteoblasts by modulating the activity of their receptors in genetically engineered mice. They found a specific part of the PTH type 1 receptor has a phosphate group added to it by the IGF-1 receptor. This chemical tagging leads to changes in the cytoskeleton of osteoblasts that enhance the formation of mature bone cells known as osteocytes. Mice without this PTH receptor had reduced numbers of osteocytes in their bone. The findings reveal a novel signaling mechanism behind this cellular transition during bone building.
Collapse
|
28
|
Xu X, Zheng L, Yuan Q, Zhen G, Crane JL, Zhou X, Cao X. Transforming growth factor-β in stem cells and tissue homeostasis. Bone Res 2018; 6:2. [PMID: 29423331 PMCID: PMC5802812 DOI: 10.1038/s41413-017-0005-4] [Citation(s) in RCA: 239] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/12/2017] [Accepted: 11/15/2017] [Indexed: 02/05/2023] Open
Abstract
TGF-β 1-3 are unique multi-functional growth factors that are only expressed in mammals, and mainly secreted and stored as a latent complex in the extracellular matrix (ECM). The biological functions of TGF-β in adults can only be delivered after ligand activation, mostly in response to environmental perturbations. Although involved in multiple biological and pathological processes of the human body, the exact roles of TGF-β in maintaining stem cells and tissue homeostasis have not been well-documented until recent advances, which delineate their functions in a given context. Our recent findings, along with data reported by others, have clearly shown that temporal and spatial activation of TGF-β is involved in the recruitment of stem/progenitor cell participation in tissue regeneration/remodeling process, whereas sustained abnormalities in TGF-β ligand activation, regardless of genetic or environmental origin, will inevitably disrupt the normal physiology and lead to pathobiology of major diseases. Modulation of TGF-β signaling with different approaches has proven effective pre-clinically in the treatment of multiple pathologies such as sclerosis/fibrosis, tumor metastasis, osteoarthritis, and immune disorders. Thus, further elucidation of the mechanisms by which TGF-β is activated in different tissues/organs and how targeted cells respond in a context-dependent way can likely be translated with clinical benefits in the management of a broad range of diseases with the involvement of TGF-β.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Gehua Zhen
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Janet L. Crane
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
29
|
Xie Z, Weng S, Li H, Yu X, Lu S, Huang K, Wu Z, Bai B, Boodhun V, Yang L. Teriparatide promotes healing of critical size femur defect through accelerating angiogenesis and degradation of β-TCP in OVX osteoporotic rat model. Biomed Pharmacother 2017; 96:960-967. [DOI: 10.1016/j.biopha.2017.11.141] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/21/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
|
30
|
Poudel SB, Bhattarai G, Kook SH, Shin YJ, Kwon TH, Lee SY, Lee JC. Recombinant human IGF-1 produced by transgenic plant cell suspension culture enhances new bone formation in calvarial defects. Growth Horm IGF Res 2017; 36:1-10. [PMID: 28787635 DOI: 10.1016/j.ghir.2017.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 07/20/2017] [Accepted: 07/28/2017] [Indexed: 12/16/2022]
Abstract
Transgenic plant cell suspension culture systems have been utilized extensively as convenient and efficient expression systems for the production of recombinant human growth factors. We produced insulin-like growth factor-1 using a plant suspension culture system (p-IGF-1) and explored its effect on new bone formation in calvarial defects. We also compared the bone regenerating potential of p-IGF-1 with commercial IGF-1 derived from Escherichia coli (e-IGF-1). Male C57BL/6 mice underwent calvarial defect surgery, and the defects were loaded with absorbable collagen sponge (ACS) only (ACS group) or ACS impregnated with 13μg of p-IGF-1 (p-IGF-1 group) or e-IGF-1 (e-IGF-1 group). The sham group did not receive any treatment with ACS or IGFs after surgery. Live μCT and histological analyses showed critical-sized bone defects in the sham group, whereas greater bone formation was observed in the p-IGF-1 and e-IGF-1 groups than the ACS group both 5 and 10weeks after surgery. Bone mineral density, bone volume, and bone surface values were also higher in the IGF groups than in the ACS group. Local delivery of p-IGF-1 or e-IGF-1 more greatly enhanced the expression of osteoblast-specific markers, but inhibited osteoclast formation, in newly formed bone compared with ACS control group. Specifically, p-IGF-1 treatment induced higher expression of alkaline phosphatase, osteocalcin, and osteopontin in the defect site than did e-IGF-1. Furthermore, treatment with p-IGF-1, but not e-IGF-1, increased mineralization of MC3T3-E1 cells, with the attendant upregulation of osteogenic marker genes. Collectively, our findings suggest the potential of p-IGF-1 in promoting the processes required for bone regeneration.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- Cluster for Craniofacial Development & Regeneration Research, Institute of Oral Biosciences, Chonbuk National University, Jeonju 54896, South Korea
| | - Govinda Bhattarai
- Cluster for Craniofacial Development & Regeneration Research, Institute of Oral Biosciences, Chonbuk National University, Jeonju 54896, South Korea
| | - Sung-Ho Kook
- Cluster for Craniofacial Development & Regeneration Research, Institute of Oral Biosciences, Chonbuk National University, Jeonju 54896, South Korea; Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Chonbuk National University, Jeonju 54896, South Korea
| | - Yun-Ji Shin
- Natural Bio-Materials Inc., Iksan 54631, South Korea
| | - Tae-Ho Kwon
- Natural Bio-Materials Inc., Iksan 54631, South Korea
| | - Seung-Youp Lee
- Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju 54896, South Korea.
| | - Jeong-Chae Lee
- Cluster for Craniofacial Development & Regeneration Research, Institute of Oral Biosciences, Chonbuk National University, Jeonju 54896, South Korea; Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Chonbuk National University, Jeonju 54896, South Korea.
| |
Collapse
|
31
|
Corrado A, Sanpaolo ER, Di Bello S, Cantatore FP. Osteoblast as a target of anti-osteoporotic treatment. Postgrad Med 2017; 129:858-865. [PMID: 28770650 DOI: 10.1080/00325481.2017.1362312] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Osteoblasts are mesenchymal cells that play a key role in maintaining bone homeostasis; they are responsible for the production of extracellular matrix proteins, regulation of matrix mineralization, control of bone remodeling and regulate osteoclast differentiation. Osteoblasts have an essential role in the pathogenesis of many bone diseases, particularly osteoporosis. For many decades, the main current available treatments for osteoporosis have been represented by anti-resorptive drugs, such as bisphosphonates, which act mainly by inhibiting osteoclasts maturation, proliferation and activity; nevertheless, in recent years much attention has been paid on anabolic aspects of osteoporosis treatment. Many experimental evidences support the hypothesis of direct effects of the classical anti-resorptive drugs also on osteoblasts, and recent progress in understanding bone physiology have led to the development of new pharmacological agents such as anti-sclerostin antibodies and teriparatide which directly target osteoblasts, inducing anabolic effects and promoting bone formation.
Collapse
Affiliation(s)
- Addolorata Corrado
- a Rheumatology Clinic Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Eliana Rita Sanpaolo
- a Rheumatology Clinic Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Silvana Di Bello
- a Rheumatology Clinic Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Francesco Paolo Cantatore
- a Rheumatology Clinic Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| |
Collapse
|
32
|
Zhou S, Glowacki J. Chronic kidney disease and vitamin D metabolism in human bone marrow-derived MSCs. Ann N Y Acad Sci 2017; 1402:43-55. [PMID: 28926112 PMCID: PMC5659722 DOI: 10.1111/nyas.13464] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/09/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022]
Abstract
Vitamin D that is synthesized in the skin or is ingested undergoes sequential steps of metabolic activation via a cascade of cytochrome P450 enzymatic hydroxylations in the liver and kidney to produce 1α,25-dihydroxyvitamin D (1α,25(OH)2 D). There are many tissues that are able to synthesize 1α,25(OH)2 D, but the biological significance of extrarenal hydroxylases is unresolved. Human marrow-derived mesenchymal stem cells (marrow stromal cells, hMSCs) give rise to osteoblasts, and their differentiation is stimulated by 1α,25(OH)2 D. In addition to being targets of 1α,25(OH)2 D, hMSCs can synthesize it; on the basis of those observations, we further examined the local autocrine/paracrine role of vitamin D metabolism in osteoblast differentiation. Research with hMSCs from well-characterized subjects provides an innovative opportunity to evaluate the effects of clinical attributes on the regulation of hMSCs. Like the renal 1α-hydroxylase, the enzyme in hMSCs is constitutively decreased with age and chronic kidney disease (CKD); both are regulated by PTH1-34, insulin-like growth factor 1, calcium, 1α,25(OH)2 D, 25(OH)D, and fibroblast growth factor 23. CKD is associated with impaired renal biosynthesis of 1α,25(OH)2 D, low bone mass, and increased fracture risk. Studies with hMSCs from CKD patients or aged subjects indicate that circulating 25(OH)D may have an important role in osteoblast differentiation on vitamin D metabolism and action in hMSCs.
Collapse
Affiliation(s)
- Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Julie Glowacki
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
33
|
Lee WC, Guntur AR, Long F, Rosen CJ. Energy Metabolism of the Osteoblast: Implications for Osteoporosis. Endocr Rev 2017; 38:255-266. [PMID: 28472361 PMCID: PMC5460680 DOI: 10.1210/er.2017-00064] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/25/2017] [Indexed: 01/14/2023]
Abstract
Osteoblasts, the bone-forming cells of the remodeling unit, are essential for growth and maintenance of the skeleton. Clinical disorders of substrate availability (e.g., diabetes mellitus, anorexia nervosa, and aging) cause osteoblast dysfunction, ultimately leading to skeletal fragility and osteoporotic fractures. Conversely, anabolic treatments for osteoporosis enhance the work of the osteoblast by altering osteoblast metabolism. Emerging evidence supports glycolysis as the major metabolic pathway to meet ATP demand during osteoblast differentiation. Glut1 and Glut3 are the principal transporters of glucose in osteoblasts, although Glut4 has also been implicated. Wnt signaling induces osteoblast differentiation and activates glycolysis through mammalian target of rapamycin, whereas parathyroid hormone stimulates glycolysis through induction of insulin-like growth factor-I. Glutamine is an alternate fuel source for osteogenesis via the tricarboxylic acid cycle, and fatty acids can be metabolized to generate ATP via oxidative phosphorylation although temporal specificity has not been established. More studies with new model systems are needed to fully understand how the osteoblast utilizes fuel substrates in health and disease and how that impacts metabolic bone diseases.
Collapse
Affiliation(s)
- Wen-Chih Lee
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Anyonya R Guntur
- Maine Medical Center Research Institute, Scarborough, Maine 04074
| | - Fanxin Long
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110.,Departments of Medicine and Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Scarborough, Maine 04074
| |
Collapse
|
34
|
PTH[1-34] improves the effects of core decompression in early-stage steroid-associated osteonecrosis model by enhancing bone repair and revascularization. PLoS One 2017; 12:e0178781. [PMID: 28562696 PMCID: PMC5451136 DOI: 10.1371/journal.pone.0178781] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/18/2017] [Indexed: 12/31/2022] Open
Abstract
Steroid-associated osteonecrosis (SAON) might induce bone collapse and subsequently lead to joint arthroplasty. Core decompression (CD) is regarded as an effective therapy for early-stage SAON, but the prognosis is unsatisfactory due to incomplete bone repair. Parathyroid hormone[1–34] (PTH[1–34]) has demonstrated positive efficacy in promoting bone formation. We therefore evaluated the effects of PTH on improving the effects of CD in Early-Stage SAON. Distal femoral CD was performed two weeks after osteonecrosis induction or vehicle injection, with ten of the ON-induced rabbits being subjected to six-week PTH[1–34] treatment and the others, including ON-induced and non-induced rabbits, being treated with vehicle. MRI confirmed that intermittent PTH administration improved SAON after CD therapy. Micro-CT showed increased bone formation within the tunnel. Bone repair was enhanced with decreased empty osteocyte lacunae and necrosis foci area, resulting in enhanced peak load and stiffness of the tunnel. Additionally, PTH enlarged the mean diameter of vessels in the marrow and increased the number of vessels within the tunnels, as well as elevated the expression of BMP-2, RUNX2, IGF-1, bFGF and VEGF, together with serum OCN and VEGF levels. Therefore, PTH[1–34] enhances the efficacy of CD on osteogenesis and neovascularization, thus promoting bone and blood vessels repair in the SAON model.
Collapse
|
35
|
Davies OG, Liu Y, Player DJ, Martin NRW, Grover LM, Lewis MP. Defining the Balance between Regeneration and Pathological Ossification in Skeletal Muscle Following Traumatic Injury. Front Physiol 2017; 8:194. [PMID: 28421001 PMCID: PMC5376571 DOI: 10.3389/fphys.2017.00194] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/15/2017] [Indexed: 12/15/2022] Open
Abstract
Heterotopic ossification (HO) is characterized by the formation of bone at atypical sites. This type of ectopic bone formation is most prominent in skeletal muscle, most frequently resulting as a consequence of physical trauma and associated with aberrant tissue regeneration. The condition is debilitating, reducing a patient's range of motion and potentially causing severe pathologies resulting from nerve and vascular compression. Despite efforts to understand the pathological processes governing HO, there remains a lack of consensus regarding the micro-environmental conditions conducive to its formation, and attempting to define the balance between muscle regeneration and pathological ossification remains complex. The development of HO is thought to be related to a complex interplay between factors released both locally and systemically in response to trauma. It develops as skeletal muscle undergoes significant repair and regeneration, and is likely to result from the misdirected differentiation of endogenous or systemically derived progenitors in response to biochemical and/or environmental cues. The process can be sequentially delineated by the presence of inflammation, tissue breakdown, adipogenesis, hypoxia, neo-vasculogenesis, chondrogenesis and ossification. However, exactly how each of these stages contributes to the formation of HO is at present not well understood. Our previous review examined the cellular contribution to HO. Therefore, the principal aim of this review will be to comprehensively outline changes in the local tissue micro-environment following trauma, and identify how these changes can alter the balance between skeletal muscle regeneration and ectopic ossification. An understanding of the mechanisms governing this condition is required for the development and advancement of HO prophylaxis and treatment, and may even hold the key to unlocking novel methods for engineering hard tissues.
Collapse
Affiliation(s)
- Owen G Davies
- School of Sport, Exercise and Health Sciences, Loughborough UniversityLoughborough, UK.,School of Chemical Engineering, University of BirminghamBirmingham, UK
| | - Yang Liu
- Wolfson School of Mechanical and Manufacturing Engineering, Loughborough UniversityLoughborough, UK
| | - Darren J Player
- School of Sport, Exercise and Health Sciences, Loughborough UniversityLoughborough, UK
| | - Neil R W Martin
- School of Sport, Exercise and Health Sciences, Loughborough UniversityLoughborough, UK
| | - Liam M Grover
- School of Chemical Engineering, University of BirminghamBirmingham, UK
| | - Mark P Lewis
- National Centre for Sport and Exercise Medicine, Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, School of Sport, Exercise and Health Sciences, Loughborough UniversityLoughborough, UK
| |
Collapse
|
36
|
Siddiqui JA, Partridge NC. Physiological Bone Remodeling: Systemic Regulation and Growth Factor Involvement. Physiology (Bethesda) 2017; 31:233-45. [PMID: 27053737 DOI: 10.1152/physiol.00061.2014] [Citation(s) in RCA: 253] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Bone remodeling is essential for adult bone homeostasis. It comprises two phases: bone formation and resorption. The balance between the two phases is crucial for sustaining bone mass and systemic mineral homeostasis. This review highlights recent work on physiological bone remodeling and discusses our knowledge of how systemic and growth factors regulate this process.
Collapse
Affiliation(s)
- Jawed A Siddiqui
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - Nicola C Partridge
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| |
Collapse
|
37
|
Abstract
Growth hormone (GH) exerts several effects on the skeleton, mediated either directly or indirectly, leading to increased bone formation and resorption rates. Patients with growth hormone deficiency (GHD) of adult onset have decreased bone mineral density (BMD) and increased fracture risk. Some, but not all, studies have found that adults with childhood onset GHD also have lower BMD than healthy controls. Adults with GHD of childhood onset have smaller bone dimensions, leading to possible underestimation of areal BMD (measured by dual energy X-ray absorptiometry), thus potentially confounding the interpretation of densitometric data. Available data suggest that patients with childhood onset GHD are at increased fracture risk. Prospective studies and some clinical trials found that GH replacement for at least 18-24 months leads to increased BMD. Retrospective and prospective data suggest that GH replacement is associated with decreased fracture risk in adults. However, data from randomized clinical trials are lacking.
Collapse
Affiliation(s)
- Nicholas A Tritos
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Wippert PM, Rector M, Kuhn G, Wuertz-Kozak K. Stress and Alterations in Bones: An Interdisciplinary Perspective. Front Endocrinol (Lausanne) 2017; 8:96. [PMID: 28507534 PMCID: PMC5410657 DOI: 10.3389/fendo.2017.00096] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/18/2017] [Indexed: 11/29/2022] Open
Abstract
Decades of research have demonstrated that physical stress (PS) stimulates bone remodeling and affects bone structure and function through complex mechanotransduction mechanisms. Recent research has laid ground to the hypothesis that mental stress (MS) also influences bone biology, eventually leading to osteoporosis and increased bone fracture risk. These effects are likely exerted by modulation of hypothalamic-pituitary-adrenal axis activity, resulting in an altered release of growth hormones, glucocorticoids and cytokines, as demonstrated in human and animal studies. Furthermore, molecular cross talk between mental and PS is thought to exist, with either synergistic or preventative effects on bone disease progression depending on the characteristics of the applied stressor. This mini review will explain the emerging concept of MS as an important player in bone adaptation and its potential cross talk with PS by summarizing the current state of knowledge, highlighting newly evolving notions (such as intergenerational transmission of stress and its epigenetic modifications affecting bone) and proposing new research directions.
Collapse
Affiliation(s)
- Pia-Maria Wippert
- Department of Health Sciences, Institute of Sociology of Health and Physical Activity, University of Potsdam, Potsdam, Germany
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
- *Correspondence: Pia-Maria Wippert,
| | - Michael Rector
- Department of Health Sciences, Institute of Sociology of Health and Physical Activity, University of Potsdam, Potsdam, Germany
| | - Gisela Kuhn
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Karin Wuertz-Kozak
- Department of Health Sciences, Institute of Sociology of Health and Physical Activity, University of Potsdam, Potsdam, Germany
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
- Schön Klinik München Harlaching, Munich, Germany
- Spine Center, Academic Teaching Hospital and Spine Research Institute, Paracelsus Private Medical University Salzburg, Salzburg, Austria
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Armakolas N, Armakolas A, Antonopoulos A, Dimakakos A, Stathaki M, Koutsilieris M. The role of the IGF-1 Ec in myoskeletal system and osteosarcoma pathophysiology. Crit Rev Oncol Hematol 2016; 108:137-145. [DOI: 10.1016/j.critrevonc.2016.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 10/05/2016] [Accepted: 11/13/2016] [Indexed: 11/28/2022] Open
|
40
|
Khan MP, Khan K, Yadav PS, Singh AK, Nag A, Prasahar P, Mittal M, China SP, Tewari MC, Nagar GK, Tewari D, Trivedi AK, Sanyal S, Bandyopadhyay A, Chattopadhyay N. BMP signaling is required for adult skeletal homeostasis and mediates bone anabolic action of parathyroid hormone. Bone 2016; 92:132-144. [PMID: 27567726 DOI: 10.1016/j.bone.2016.08.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 08/15/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
Abstract
Bmp2 and Bmp4 genes were ablated in adult mice (KO) using a conditional gene knockout technology. Bones were evaluated by microcomputed tomography (μCT), bone strength tester, histomorphometry and serum biochemical markers of bone turnover. Drill-hole was made at femur metaphysis and bone regeneration in the hole site was measured by calcein binding and μCT. Mice were either sham operated (ovary intact) or ovariectomized (OVX), and treated with human parathyroid hormone (PTH), 17β-estradiol (E2) or vehicle. KO mice displayed trabecular bone loss, diminished osteoid formation and reduced biomechanical strength compared with control (expressing Bmp2 and Bmp4). Both osteoblast and osteoclast functions were impaired in KO mice. Bone histomorphomtery and serum parameters established a low turnover bone loss in KO mice. Bone regeneration at the drill-hole site in KO mice was lower than control. However, deletion of Bmp2 gene alone had no effect on skeleton, an outcome similar to that reported previously for deletion of Bmp4 gene. Both PTH and E2 resulted in skeletal preservation in control-OVX, whereas in KO-OVX, E2 but not PTH was effective which suggested that the skeletal action of PTH required Bmp ligands but E2 did not. To determine cellular effects of Bmp2 and Bmp4, we used bone marrow stromal cells in which PTH but not E2 stimulated both Bmp2 and Bmp4 synthesis leading to increased Smad1/5 phosphorylation. Taken together, we conclude that Bmp2 and Bmp4 are essential for maintaining adult skeletal homeostasis and mediating the anabolic action of PTH.
Collapse
Affiliation(s)
- Mohd Parvez Khan
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Kainat Khan
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Prem Swaroop Yadav
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur 208016, India
| | - Abhishek Kumar Singh
- Division of Biochemistry, CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Aditi Nag
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur 208016, India
| | - Paritosh Prasahar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur 208016, India
| | - Monika Mittal
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India; AcSIR, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shyamsundar Pal China
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India; AcSIR, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Mahesh Chandra Tewari
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Geet Kumar Nagar
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Deepshikha Tewari
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Arun Kumar Trivedi
- Division of Biochemistry, CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Sabyasachi Sanyal
- Division of Biochemistry, CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur 208016, India.
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India; AcSIR, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
41
|
Devesa J, Almengló C, Devesa P. Multiple Effects of Growth Hormone in the Body: Is it Really the Hormone for Growth? Clin Med Insights Endocrinol Diabetes 2016; 9:47-71. [PMID: 27773998 PMCID: PMC5063841 DOI: 10.4137/cmed.s38201] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/12/2016] [Accepted: 09/19/2016] [Indexed: 12/17/2022] Open
Abstract
In this review, we analyze the effects of growth hormone on a number of tissues and organs and its putative role in the longitudinal growth of an organism. We conclude that the hormone plays a very important role in maintaining the homogeneity of tissues and organs during the normal development of the human body or after an injury. Its effects on growth do not seem to take place during the fetal period or during the early infancy and are mediated by insulin-like growth factor I (IGF-I) during childhood and puberty. In turn, IGF-I transcription is dependent on an adequate GH secretion, and in many tissues, it occurs independent of GH. We propose that GH may be a prohormone, rather than a hormone, since in many tissues and organs, it is proteolytically cleaved in a tissue-specific manner giving origin to shorter GH forms whose activity is still unknown.
Collapse
Affiliation(s)
- Jesús Devesa
- Scientific Direction, Medical Center Foltra, Teo, Spain
| | | | - Pablo Devesa
- Research and Development, Medical Center Foltra, 15886-Teo, Spain
| |
Collapse
|
42
|
Ahi EP. Signalling pathways in trophic skeletal development and morphogenesis: Insights from studies on teleost fish. Dev Biol 2016; 420:11-31. [PMID: 27713057 DOI: 10.1016/j.ydbio.2016.10.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/02/2016] [Accepted: 10/03/2016] [Indexed: 12/12/2022]
Abstract
During the development of the vertebrate feeding apparatus, a variety of complicated cellular and molecular processes participate in the formation and integration of individual skeletal elements. The molecular mechanisms regulating the formation of skeletal primordia and their development into specific morphological structures are tightly controlled by a set of interconnected signalling pathways. Some of these pathways, such as Bmp, Hedgehog, Notch and Wnt, are long known for their pivotal roles in craniofacial skeletogenesis. Studies addressing the functional details of their components and downstream targets, the mechanisms of their interactions with other signals as well as their potential roles in adaptive morphological divergence, are currently attracting considerable attention. An increasing number of signalling pathways that had previously been described in different biological contexts have been shown to be important in the regulation of jaw skeletal development and morphogenesis. In this review, I provide an overview of signalling pathways involved in trophic skeletogenesis emphasizing studies of the most species-rich group of vertebrates, the teleost fish, which through their evolutionary history have undergone repeated episodes of spectacular trophic diversification.
Collapse
Affiliation(s)
- Ehsan Pashay Ahi
- Institute of Zoology, University of Graz, Universitätsplatz 2, A-8010 Graz, Austria; Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland.
| |
Collapse
|
43
|
Lindsey RC, Mohan S. Skeletal effects of growth hormone and insulin-like growth factor-I therapy. Mol Cell Endocrinol 2016; 432:44-55. [PMID: 26408965 PMCID: PMC4808510 DOI: 10.1016/j.mce.2015.09.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 10/23/2022]
Abstract
The growth hormone/insulin-like growth factor (GH/IGF) axis is critically important for the regulation of bone formation, and deficiencies in this system have been shown to contribute to the development of osteoporosis and other diseases of low bone mass. The GH/IGF axis is regulated by a complex set of hormonal and local factors which can act to regulate this system at the level of the ligands, receptors, IGF binding proteins (IGFBPs), or IGFBP proteases. A combination of in vitro studies, transgenic animal models, and clinical human investigations has provided ample evidence of the importance of the endocrine and local actions of both GH and IGF-I, the two major components of the GH/IGF axis, in skeletal growth and maintenance. GH- and IGF-based therapies provide a useful avenue of approach for the prevention and treatment of diseases such as osteoporosis.
Collapse
Affiliation(s)
- Richard C Lindsey
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA; Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA; Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA.
| |
Collapse
|
44
|
Abstract
Skeletal muscle and bone rely on a number of growth factors to undergo development, modulate growth, and maintain physiological strength. A major player in these actions is insulin-like growth factor I (IGF-I). However, because this growth factor can directly enhance muscle mass and bone density, it alters the state of the musculoskeletal system indirectly through mechanical crosstalk between these two organ systems. Thus, there are clearly synergistic actions of IGF-I that extend beyond the direct activity through its receptor. This review will cover the production and signaling of IGF-I as it pertains to muscle and bone, the chemical and mechanical influences that arise from IGF-I activity, and the potential for therapeutic strategies based on IGF-I. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
|
45
|
Tritos NA, Klibanski A. Effects of Growth Hormone on Bone. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 138:193-211. [PMID: 26940392 DOI: 10.1016/bs.pmbts.2015.10.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Describe the effects of growth hormone (GH) and insulin-like growth factor 1 (IGF-1) on the skeleton. FINDINGS The GH and IGF-1 axis has pleiotropic effects on the skeleton throughout the lifespan by influencing bone formation and resorption. GH deficiency leads to decreased bone turnover, delayed statural growth in children, low bone mass, and increased fracture risk in adults. GH replacement improves adult stature in GH deficient children, increases bone mineral density (BMD) in adults, and helps to optimize peak bone acquisition in patients, during the transition from adolescence to adulthood, who have persistent GH deficiency. Observational studies suggest that GH replacement may mitigate the excessive fracture risk associated with GH deficiency. Acromegaly, a state of GH and IGF-1 excess, is associated with increased bone turnover and decreased BMD in the lumbar spine observed in some studies, particularly in patients with hypogonadism. In addition, patients with acromegaly appear to be at an increased risk of morphometric-vertebral fractures, especially in the presence of active disease or concurrent hypogonadism. GH therapy also has beneficial effects on statural growth in several conditions characterized by GH insensitivity, including chronic renal failure, Turner syndrome, Prader-Willi syndrome, postnatal growth delay in patients with intrauterine growth retardation who do not demonstrate catchup growth, idiopathic short stature, short stature homeobox-containing (SHOX) gene mutations, and Noonan syndrome. SUMMARY GH and IGF-1 have important roles in skeletal physiology, and GH has an important therapeutic role in both GH deficiency and insensitivity states.
Collapse
Affiliation(s)
- Nicholas A Tritos
- Neuroendocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA.
| | - Anne Klibanski
- Neuroendocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Xi G, Rosen CJ, Clemmons DR. IGF-I and IGFBP-2 Stimulate AMPK Activation and Autophagy, Which Are Required for Osteoblast Differentiation. Endocrinology 2016; 157:268-81. [PMID: 26556533 PMCID: PMC4701891 DOI: 10.1210/en.2015-1690] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/06/2015] [Indexed: 12/13/2022]
Abstract
IGF-I/insulin-like growth factor binding protein 2 (IGFBP-2) coordinately stimulate osteoblast differentiation but the mechanisms by which they function have not been determined. AMP-activated protein kinase (AMPK) is induced during differentiation and AMPK knockout mice have reduced bone mass. IGF-I modulates AMPK in other cell types; therefore, these studies determined whether IGF-I/IGFBP-2 stimulate AMPK activation and the mechanism by which AMPK modulates differentiation. Calvarial osteoblasts and MC-3T3 cells expressed activated AMPK early in differentiation and AMPK inhibitors attenuated differentiation. However, expression of constitutively activated AMPK inhibited differentiation. To resolve this discrepancy we analyzed the time course of AMPK induction. AMPK activation was required early in differentiation (day 3-6) but down-regulation of AMPK after day 9 was also necessary. IGF-I/IGFBP-2 induced AMPK through their respective receptors and blocking-receptor activation blocked AMPK induction. To determine the mechanism by which AMPK functioned we analyzed components of the autophagosome. Activated AMPK stimulated ULK-1 S555 phosphorylation as well as beclin-1 and microtubule-associated protein 1A/1B light-chain phosphatidylethanolamine conjugate (LC3II) induction. Inhibition of AMPK attenuated these changes and direct inhibition of autophagy inhibited differentiation. Conversely, expression of activated AMPK was associated with persistence of these changes beyond day 9 and inhibited differentiation. Blocking AMPK activation after day 9 down-regulated these autophagosome components and rescued differentiation. This allowed induction of mechanistic target of rapamycin and AKT, which suppressed autophagy. The results show that early induction of AMPK in response to IGF-I/IGFBP-2 followed by suppression is required for osteoblast differentiation. AMPK functions through stimulation of autophagy. The findings suggest that these early catabolic changes are important for determining the energy source for osteoblast respiration and down-regulation of these components may be required for induction of glycolysis, which is required during the final anabolic stages of differentiation.
Collapse
Affiliation(s)
- Gang Xi
- Department of Medicine (G.X., D.R.C.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599; and Maine Medical Center Research Institute (C.J.R.), Scarborough, Maine 04074
| | - Clifford J Rosen
- Department of Medicine (G.X., D.R.C.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599; and Maine Medical Center Research Institute (C.J.R.), Scarborough, Maine 04074
| | - David R Clemmons
- Department of Medicine (G.X., D.R.C.), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599; and Maine Medical Center Research Institute (C.J.R.), Scarborough, Maine 04074
| |
Collapse
|
47
|
Wang Y, Menendez A, Fong C, ElAlieh HZ, Kubota T, Long R, Bikle DD. IGF-I Signaling in Osterix-Expressing Cells Regulates Secondary Ossification Center Formation, Growth Plate Maturation, and Metaphyseal Formation During Postnatal Bone Development. J Bone Miner Res 2015; 30:2239-48. [PMID: 26011431 PMCID: PMC9042009 DOI: 10.1002/jbmr.2563] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 05/01/2015] [Accepted: 05/15/2015] [Indexed: 12/28/2022]
Abstract
To investigate the role of IGF-I signaling in osterix (OSX)-expressing cells in the skeleton, we generated IGF-I receptor (IGF-IR) knockout mice ((OSX)IGF-IRKO) (floxed-IGF-IR mice × OSX promoter-driven GFP-labeled cre-recombinase [(OSX)GFPcre]), and monitored postnatal bone development. At day 2 after birth (P2), (OSX)GFP-cre was highly expressed in the osteoblasts in the bone surface of the metaphysis and in the prehypertrophic chondrocytes (PHCs) and inner layer of perichondral cells (IPCs). From P7, (OSX)GFP-cre was highly expressed in PHCs, IPCs, cartilage canals (CCs), and osteoblasts (OBs) in the epiphyseal secondary ossification center (SOC), but was only slightly expressed in the OBs in the metaphysis. Compared with the control mice, the IPC proliferation was decreased in the (OSX)IGF-IRKOs. In these mice, fewer IPCs invaded into the cartilage, resulting in delayed formation of the CC and SOC. Immunohistochemistry indicated a reduction of vessel number and lower expression of VEGF and ephrin B2 in the IPCs and SOC of (OSX)IGF-IRKOs. Quantitative real-time PCR revealed that the mRNA levels of the matrix degradation markers, MMP-9, 13 and 14, were decreased in the (OSX)IGF-IRKOs compared with the controls. The (OSX)IGF-IRKO also showed irregular morphology of the growth plate and less trabecular bone in the tibia and femur from P7 to 7 weeks, accompanied by decreased chondrocyte proliferation, altered chondrocyte differentiation, and decreased osteoblast differentiation. Our data indicate that during postnatal bone development, IGF-I signaling in OSX-expressing IPCs promotes IPC proliferation and cartilage matrix degradation and increases ephrin B2 production to stimulate vascular endothelial growth factor (VEGF) expression and vascularization. These processes are required for normal CC formation in the establishment of the SOC. Moreover, IGF-I signaling in the OSX-expressing PHC is required for growth plate maturation and osteoblast differentiation in the development of the metaphysis.
Collapse
Affiliation(s)
- Yongmei Wang
- Endocrine Unit, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Alicia Menendez
- Endocrine Unit, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Chak Fong
- Endocrine Unit, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Hashem Z ElAlieh
- Endocrine Unit, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Takuo Kubota
- Endocrine Unit, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Roger Long
- Endocrine Unit, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Daniel D Bikle
- Endocrine Unit, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, CA, USA
| |
Collapse
|
48
|
Clifton KB, Conover CA. Pregnancy-associated plasma protein-A modulates the anabolic effects of parathyroid hormone in mouse bone. Bone 2015; 81:413-416. [PMID: 26297833 PMCID: PMC4641039 DOI: 10.1016/j.bone.2015.08.015] [Citation(s) in RCA: 5] [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] [Received: 01/31/2015] [Revised: 08/10/2015] [Accepted: 08/17/2015] [Indexed: 11/27/2022]
Abstract
Intermittent parathyroid hormone (PTH) is a potent anabolic therapy for bone, and several studies have implicated local insulin-like growth factor (IGF) signaling in mediating this effect. The IGF system is complex and includes ligands and receptors, as well as IGF binding proteins (IGFBPs) and IGFBP proteases. Pregnancy-associated plasma protein-A (PAPP-A) is a metalloprotease expressed by osteoblasts in vitro that has been shown to enhance local IGF action through cleavage of inhibitory IGFBP-4. This study was set up to test two specific hypotheses: 1) Intermittent PTH treatment increases the expression of IGF-I, IGFBP-4 and PAPP-A in bone in vivo, thereby increasing local IGF activity. 2) In the absence of PAPP-A, local IGF activity and the anabolic effects of PTH on bone are reduced. Wild-type (WT) and PAPP-A knock-out (KO) mice were treated with 80 μg/kg human PTH 1-34 or vehicle by subcutaneous injection five days per week for six weeks. IGF-I, IGFBP-4 and PAPP-A mRNA expression in bone were significantly increased in response to PTH treatment. PTH treatment of WT mice, but not PAPP-A KO mice, significantly increased expression of an IGF-responsive gene. Bone mineral density (BMD), as measured by DEXA, was significantly decreased in femurs of PAPP-A KO compared to WT mice with PTH treatment. Volumetric BMD, as measured by pQCT, was significantly decreased in femoral midshaft (primarily cortical bone), but not metaphysis (primarily trabecular bone), of PAPP-A KO compared to WT mice with PTH treatment. These data suggest that stimulation of PAPP-A expression by intermittent PTH treatment contributes to PTH bone anabolism in mice.
Collapse
Affiliation(s)
- Kari B Clifton
- Division of Endocrinology, Mayo Clinic, 200 First Street, SW, Rochester, MN 55905, United States
| | - Cheryl A Conover
- Division of Endocrinology, Mayo Clinic, 200 First Street, SW, Rochester, MN 55905, United States.
| |
Collapse
|
49
|
Bikle DD, Tahimic C, Chang W, Wang Y, Philippou A, Barton ER. Role of IGF-I signaling in muscle bone interactions. Bone 2015; 80:79-88. [PMID: 26453498 PMCID: PMC4600536 DOI: 10.1016/j.bone.2015.04.036] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/11/2015] [Accepted: 04/22/2015] [Indexed: 12/16/2022]
Abstract
Skeletal muscle and bone rely on a number of growth factors to undergo development, modulate growth, and maintain physiological strength. A major player in these actions is insulin-like growth factor I (IGF-I). However, because this growth factor can directly enhance muscle mass and bone density, it alters the state of the musculoskeletal system indirectly through mechanical crosstalk between these two organ systems. Thus, there are clearly synergistic actions of IGF-I that extend beyond the direct activity through its receptor. This review will cover the production and signaling of IGF-I as it pertains to muscle and bone, the chemical and mechanical influences that arise from IGF-I activity, and the potential for therapeutic strategies based on IGF-I. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Daniel D Bikle
- VA Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Candice Tahimic
- VA Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Wenhan Chang
- VA Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Yongmei Wang
- VA Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Anastassios Philippou
- National and Kapodistrian University of Athens, Department of Physiology, Medical School, Goudi-Athens, Greece
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
50
|
Riddle RC. Parathyroid Hormone Reprograms Osteoblast Metabolism. J Bone Miner Res 2015; 30:1956-8. [PMID: 26448640 PMCID: PMC4824191 DOI: 10.1002/jbmr.2727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 10/04/2015] [Accepted: 10/06/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Ryan C Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| |
Collapse
|