1
|
Zoccali C, Vervloet MG, Evenepoel P, Massy Z, Cozzolino M, Mallamaci F, Lederer ED, Andia JC, Drueke TB. The autonomic nervous system and bone health in chronic kidney disease. Eur J Clin Invest 2025; 55:e70007. [PMID: 39985733 DOI: 10.1111/eci.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/12/2025] [Indexed: 02/24/2025]
Abstract
Besides the well-known role of hormonal factors in mineral and bone metabolism, the sympathetic nervous system participates in this regulation by inhibiting bone formation and promoting bone resorption, primarily via β-adrenergic receptors expressed on osteoblasts. Conversely, the parasympathetic system, through cholinergic signalling, inhibits osteoclast activity, promoting bone formation and maintaining skeletal homeostasis. This review presents the role of the autonomic nervous system, with particular focus on the potential role of β-blockers, especially β1-selective blockers, in modulating bone health in people with normal kidney function and those with CKD. While early studies with non-selective β-blockers like propranolol showed mixed results, recent findings in postmenopausal women suggested that β1-selective β-blockers could enhance bone density by modulating sympathetic activity. Trial emulation using large databases and eventually randomized controlled trials are needed to test the hypothesis that β-blockade can favourably impact bone disease in patients with kidney failure.
Collapse
Affiliation(s)
- Carmine Zoccali
- Renal Research Institute, New York, New York, USA
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), Reggio di Calabria, Italy
| | - Marc G Vervloet
- Nephrology Department, Amsterdam UMC, Amsterdam, The Netherlands
| | - Pieter Evenepoel
- Department of Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Ziad Massy
- Inserm Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Sud University (UPS) and Versailles Saint-Quentin-En-Yvelines University (Paris-Ile-de-France-Ouest University, UVSQ), Villejuif, France
- Association Pour l'Utilisation du Rein Artificiel Dans la région Parisienne (AURA), Paris, France
- Department of Nephrology, Ambroise Paré University Hospital, APHP, Boulogne-Billancourt, Paris, France
| | - Mario Cozzolino
- Renal Division, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milan, Italy
| | - Francesca Mallamaci
- Unità Operativa di Nefrologia e Trapianto Renale, Grande Ospedale Metropolitano, Reggio Calabria, Italy
- Clinical Epidemiology Unit of the CNR Institute of Clinical Physiology, Grande Ospedale Metropolitan, Reggio Calabria, Italy
| | - Eleanor D Lederer
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Veterans Affairs North Texas Health Care Services, Dallas, Texas, USA
- UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jorge Cannata Andia
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), Madrid, Spain
- Department of Medicine, Universidad de Oviedo, Oviedo, Spain
| | - Tilman B Drueke
- Inserm Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Sud University (UPS), Villejuif, France
- Versailles Saint-Quentin-En-Yvelines University (Paris-Ile-de-France-Ouest University, UVSQ), Villejuif, France
- Inserm U 1038, Centre de Recherche Des Cordeliers, Paris-Cité University, Sorbonne-University, Paris, France
| |
Collapse
|
2
|
Ma C, Zhang Y, Cao Y, Hu CH, Zheng CX, Jin Y, Sui BD. Autonomic neural regulation in mediating the brain-bone axis: mechanisms and implications for regeneration under psychological stress. QJM 2024; 117:95-108. [PMID: 37252831 DOI: 10.1093/qjmed/hcad108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Indexed: 06/01/2023] Open
Abstract
Efficient regeneration of bone defects caused by disease or significant trauma is a major challenge in current medicine, which is particularly difficult yet significant under the emerging psychological stress in the modern society. Notably, the brain-bone axis has been proposed as a prominent new concept in recent years, among which autonomic nerves act as an essential and emerging skeletal pathophysiological factor related to psychological stress. Studies have established that sympathetic cues lead to impairment of bone homeostasis mainly through acting on mesenchymal stem cells (MSCs) and their derivatives with also affecting the hematopoietic stem cell (HSC)-lineage osteoclasts, and the autonomic neural regulation of stem cell lineages in bone is increasingly recognized to contribute to the bone degenerative disease, osteoporosis. This review summarizes the distribution characteristics of autonomic nerves in bone, introduces the regulatory effects and mechanisms of autonomic nerves on MSC and HSC lineages, and expounds the crucial role of autonomic neural regulation on bone physiology and pathology, which acts as a bridge between the brain and the bone. With the translational perspective, we further highlight the autonomic neural basis of psychological stress-induced bone loss and a series of pharmaceutical therapeutic strategies and implications toward bone regeneration. The summary of research progress in this field will add knowledge to the current landscape of inter-organ crosstalk and provide a medicinal basis for the achievement of clinical bone regeneration in the future.
Collapse
Affiliation(s)
- C Ma
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Y Zhang
- Department of Medical Rehabilitation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Y Cao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - C-H Hu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - C-X Zheng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Y Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - B-D Sui
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
3
|
Xu HK, Liu JX, Zheng CX, Liu L, Ma C, Tian JY, Yuan Y, Cao Y, Xing SJ, Liu SY, Li Q, Zhao YJ, Kong L, Chen YJ, Sui BD. Region-specific sympatho-adrenergic regulation of specialized vasculature in bone homeostasis and regeneration. iScience 2023; 26:107455. [PMID: 37680481 PMCID: PMC10481296 DOI: 10.1016/j.isci.2023.107455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/13/2023] [Accepted: 07/19/2023] [Indexed: 09/09/2023] Open
Abstract
Type H vessels couple angiogenesis with osteogenesis, while sympathetic cues regulate vascular and skeletal function. The crosstalk between sympathetic nerves and type H vessels in bone remains unclear. Here, we first identify close spatial connections between sympathetic nerves and type H vessels in bone, particularly in metaphysis. Sympathoexcitation, mimicked by isoproterenol (ISO) injection, reduces type H vessels and bone mass. Conversely, beta-2-adrenergic receptor (ADRB2) deficiency maintains type H vessels and bone mass in the physiological condition. In vitro experiments reveal indirect sympathetic modulation of angiogenesis via paracrine effects of mesenchymal stem cells (MSCs), which alter the transcription of multiple angiogenic genes in endothelial cells (ECs). Furthermore, Notch signaling in ECs underlies sympathoexcitation-regulated type H vessel formation, impacting osteogenesis and bone mass. Finally, propranolol (PRO) inhibits beta-adrenergic activity and protects type H vessels and bone mass against estrogen deficiency. These findings unravel the specialized neurovascular coupling in bone homeostasis and regeneration.
Collapse
Affiliation(s)
- Hao-Kun Xu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Jie-Xi Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Chen-Xi Zheng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Lu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Chao Ma
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Jiong-Yi Tian
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yuan Yuan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Exercise Immunology Center, Wuhan Sports University, Wuhan, Hubei 430079, China
| | - Yuan Cao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Shu-Juan Xing
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Si-Ying Liu
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Qiang Li
- Department of General Dentistry & Emergency, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Ya-Juan Zhao
- Department of General Dentistry & Emergency, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Liang Kong
- Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yong-Jin Chen
- Department of General Dentistry & Emergency, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Bing-Dong Sui
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| |
Collapse
|
4
|
Xiao Y, Han C, Wang Y, Zhang X, Bao R, Li Y, Chen H, Hu B, Liu S. Interoceptive regulation of skeletal tissue homeostasis and repair. Bone Res 2023; 11:48. [PMID: 37669953 PMCID: PMC10480189 DOI: 10.1038/s41413-023-00285-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 05/08/2023] [Accepted: 06/22/2023] [Indexed: 09/07/2023] Open
Abstract
Recent studies have determined that the nervous system can sense and respond to signals from skeletal tissue, a process known as skeletal interoception, which is crucial for maintaining bone homeostasis. The hypothalamus, located in the central nervous system (CNS), plays a key role in processing interoceptive signals and regulating bone homeostasis through the autonomic nervous system, neuropeptide release, and neuroendocrine mechanisms. These mechanisms control the differentiation of mesenchymal stem cells into osteoblasts (OBs), the activation of osteoclasts (OCs), and the functional activities of bone cells. Sensory nerves extensively innervate skeletal tissues, facilitating the transmission of interoceptive signals to the CNS. This review provides a comprehensive overview of current research on the generation and coordination of skeletal interoceptive signals by the CNS to maintain bone homeostasis and their potential role in pathological conditions. The findings expand our understanding of intersystem communication in bone biology and may have implications for developing novel therapeutic strategies for bone diseases.
Collapse
Affiliation(s)
- Yao Xiao
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China
| | - Changhao Han
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China
| | - Yunhao Wang
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Xinshu Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China
| | - Rong Bao
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China
| | - Yuange Li
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China
| | - Huajiang Chen
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Bo Hu
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| | - Shen Liu
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China.
| |
Collapse
|
5
|
Wank I, Niedermair T, Kronenberg D, Stange R, Brochhausen C, Hess A, Grässel S. Influence of the Peripheral Nervous System on Murine Osteoporotic Fracture Healing and Fracture-Induced Hyperalgesia. Int J Mol Sci 2022; 24:510. [PMID: 36613952 PMCID: PMC9820334 DOI: 10.3390/ijms24010510] [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] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Osteoporotic fractures are often linked to persisting chronic pain and poor healing outcomes. Substance P (SP), α-calcitonin gene-related peptide (α-CGRP) and sympathetic neurotransmitters are involved in bone remodeling after trauma and nociceptive processes, e.g., fracture-induced hyperalgesia. We aimed to link sensory and sympathetic signaling to fracture healing and fracture-induced hyperalgesia under osteoporotic conditions. Externally stabilized femoral fractures were set 28 days after OVX in wild type (WT), α-CGRP- deficient (α-CGRP -/-), SP-deficient (Tac1-/-) and sympathectomized (SYX) mice. Functional MRI (fMRI) was performed two days before and five and 21 days post fracture, followed by µCT and biomechanical tests. Sympathectomy affected structural bone properties in the fracture callus whereas loss of sensory neurotransmitters affected trabecular structures in contralateral, non-fractured bones. Biomechanical properties were mostly similar in all groups. Both nociceptive and resting-state (RS) fMRI revealed significant baseline differences in functional connectivity (FC) between WT and neurotransmitter-deficient mice. The fracture-induced hyperalgesia modulated central nociception and had robust impact on RS FC in all groups. The changes demonstrated in RS FC in fMRI might potentially be used as a bone traumata-induced biomarker regarding fracture healing under pathophysiological musculoskeletal conditions. The findings are of clinical importance and relevance as they advance our understanding of pain during osteoporotic fracture healing and provide a potential imaging biomarker for fracture-related hyperalgesia and its temporal development. Overall, this may help to reduce the development of chronic pain after fracture thereby improving the treatment of osteoporotic fractures.
Collapse
Affiliation(s)
- Isabel Wank
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Tanja Niedermair
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
| | - Daniel Kronenberg
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine (IMM), University Hospital Münster, 48149 Münster, Germany
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine (IMM), University Hospital Münster, 48149 Münster, Germany
| | | | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Susanne Grässel
- Centre for Medical Biotechnology (ZMB), Department of Orthopedic Surgery, Experimental Orthopedics, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
6
|
Targeted inhibition of osteoclastogenesis reveals the pathogenesis and therapeutics of bone loss under sympathetic neurostress. Int J Oral Sci 2022; 14:39. [PMID: 35915088 PMCID: PMC9343357 DOI: 10.1038/s41368-022-00193-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/11/2022] [Accepted: 06/23/2022] [Indexed: 12/28/2022] Open
Abstract
Sympathetic cues via the adrenergic signaling critically regulate bone homeostasis and contribute to neurostress-induced bone loss, but the mechanisms and therapeutics remain incompletely elucidated. Here, we reveal an osteoclastogenesis-centered functionally important osteopenic pathogenesis under sympatho-adrenergic activation with characterized microRNA response and efficient therapeutics. We discovered that osteoclastic miR-21 was tightly regulated by sympatho-adrenergic cues downstream the β2-adrenergic receptor (β2AR) signaling, critically modulated osteoclastogenesis in vivo by inhibiting programmed cell death 4 (Pdcd4), and mediated detrimental effects of both isoproterenol (ISO) and chronic variable stress (CVS) on bone. Intriguingly, without affecting osteoblastic bone formation, bone protection against ISO and CVS was sufficiently achieved by a (D-Asp8)-lipid nanoparticle-mediated targeted inhibition of osteoclastic miR-21 or by clinically relevant drugs to suppress osteoclastogenesis. Collectively, these results unravel a previously underdetermined molecular and functional paradigm that osteoclastogenesis crucially contributes to sympatho-adrenergic regulation of bone and establish multiple targeted therapeutic strategies to counteract osteopenias under stresses.
Collapse
|
7
|
Ehterami A, Khastar H, Soleimannejad M, Salehi M, Nazarnezhad S, Majidi Ghatar J, Bit A, JafariSani M, Abbaszadeh-Goudarzi G, Shariatifar N. Bone Regeneration in Rat using Polycaprolactone/Gelatin/Epinephrine Scaffold. Drug Dev Ind Pharm 2022; 47:1915-1923. [PMID: 35484948 DOI: 10.1080/03639045.2022.2070640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Solid supports like the extracellular matrix network are necessary for bone cell attachment and start healing in the damaged bone. Scaffolds which are made of different materials are widely used as a supportive structure in bone tissue engineering. In the current study, a 3-D Polycaprolactone/Gelatin bone scaffold was developed by blending electrospinning and freeze-drying techniques for bone tissue engineering. To improve the efficiency of the scaffold, different concentrations of epinephrine due to its effect on bone healing were loaded. Fabricated scaffolds were characterized by different tests such as surface morphology, FTIR, porosity, compressive strength, water contact angle, degradation rate. The interaction between prepared scaffolds and blood and cells was evaluated by hemolysis, and MTT test, respectively, and bone healing was evaluated by a rat calvaria defect model. Based on the results, the porosity of scaffolds was about 75% and by adding epinephrine, mechanical strength decreased while due to the hydrophilic properties of it, degradation rate increased. In vivo and in vitro studies showed the best cell proliferation and bone healing were in PCL/Gelatin/Epinephrine1%-treated group. These results showed the positive effect of fabricated scaffold on osteogenesis and bone healing and the possibility of using it in clinical trials.
Collapse
Affiliation(s)
- Arian Ehterami
- Department of Mechanical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hossein Khastar
- Department of Mechanical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran.,Sexual Health and Fertility Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.,School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mostafa Soleimannejad
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Majid Salehi
- Sexual Health and Fertility Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.,Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Simin Nazarnezhad
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jila Majidi Ghatar
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Arindam Bit
- Department of Biomedical Engineering, National Institute of Technology Raipur, India
| | - Moslem JafariSani
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ghasem Abbaszadeh-Goudarzi
- Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Nabi Shariatifar
- Department of Environmental of health engineering, school of public health, Tehran university of medical science, Tehran, Iran
| |
Collapse
|
8
|
Hu CH, Sui BD, Liu J, Dang L, Chen J, Zheng CX, Shi S, Zhao N, Dang MY, He XN, Zhang LQ, Gao PP, Chen N, Kuang HJ, Chen K, Xu XL, Yu XR, Zhang G, Jin Y. Sympathetic Neurostress Drives Osteoblastic Exosomal MiR-21 Transfer to Disrupt Bone Homeostasis and Promote Osteopenia. SMALL METHODS 2022; 6:e2100763. [PMID: 35312228 DOI: 10.1002/smtd.202100763] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/19/2021] [Indexed: 06/14/2023]
Abstract
Innervation and extracellular vesicle secretion co-exist in the local tissue microenvironment for message transfer, but whether they are interconnected to regulate organ homeostasis remains unknown. Sympatho-adrenergic activation is implicated in stress-induced depression and leads to bone loss, but the mechanisms and therapeutics are incompletely elucidated. Here, it is revealed that sympathetic neurostress through the β1/2 -adrenergic receptor (β1/2-AR) signaling triggers the transcription response of a microRNA, miR-21, in osteoblasts, which is transferred to osteoclast progenitors via exosomes for dictating osteoclastogenesis. After confirming that miR-21 deficiency retards the β1/2-AR agonist isoproterenol (ISO)-induced osteopenia, it is shown that the pharmacological inhibition of exosome release by two clinically-relevant drugs, dimethyl amiloride and omeprazole, suppresses osteoblastic miR-21 transfer and ameliorates bone loss under both ISO and chronic variable stress (CVS)-induced depression conditions. A targeted delivery approach to specifically silence osteoblastic miR-21 is further applied, which is effective in rescuing the bone remodeling balance and ameliorating ISO- and CVS-induced osteopenias. These results decipher a previously unrecognized paradigm that neural cues drive exosomal microRNA communication to regulate organ homeostasis and help to establish feasible strategies to counteract bone loss under psychological stresses.
Collapse
Affiliation(s)
- Cheng-Hu Hu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710032, China
| | - Bing-Dong Sui
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Lei Dang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Ji Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Chen-Xi Zheng
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Songtao Shi
- South China Center of Craniofacial Stem Cell Research, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Na Zhao
- Institute for Stem Cell and Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710032, China
| | - Min-Yan Dang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China
| | - Xiao-Ning He
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China
| | - Li-Qiang Zhang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China
| | - Ping-Ping Gao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China
| | - Nan Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hui-Juan Kuang
- Institute for Stem Cell and Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China
| | - Kai Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xiao-Lin Xu
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China
| | - Xiao-Rui Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710032, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China
| |
Collapse
|
9
|
Wang Z, Liu Y, Zhang J, Lin M, Xiao C, Bai H, Liu C. Mechanical loading alleviated the inhibition of β2-adrenergic receptor agonist terbutaline on bone regeneration. FASEB J 2021; 35:e22033. [PMID: 34739146 DOI: 10.1096/fj.202101045rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/14/2022]
Abstract
The long-term use of adrenergic medication in treating various conditions, such as asthma, increases the chances of bone fracture. Dynamic mechanical loading at a specific time is a method for improving bone quality and promoting healing. Therefore, we hypothesized that precisely controlling the mechanical environment can contribute to the alleviation of the negative effects of chronic treatment with the common asthma drug terbutaline, which is a β2-adrenergic receptor agonist that facilitates bone homeostasis and defect repair through its anabolic effect on osteogenic cells. Our in vitro results showed that terbutaline can directly inhibit osteogenesis by impairing osteogenic differentiation and mineralization. Chronic treatment in vivo was simulated by administering terbutaline to C57BL/6J mice for 4 weeks before bone defect surgery and mechanical loading. We utilized a stabilized tibial defect model, which allowed the application of anabolic mechanical loading. During homeostasis, chronic terbutaline treatment reduced the bone formation rate, the fracture toughness of long bones, and the concentrations of bone formation markers in the sera. During defect repair, terbutaline decreased the bone volume, type H vessel, and total blood vessel volume. Terbutaline treatment reduced the number of osteogenic cells. Periostin, which was secreted mainly by Prrx1+ osteoprogenitors and F4/80+ macrophages, was inhibited by treating the bone defect with terbutaline. Interestingly, controlled mechanical loading facilitated the recovery of bone volume and periostin expression and the number of osteogenic cells within the defect. In conclusion, mechanical loading can rescue negative effects on new bone accrual and repair induced by chronic terbutaline treatment.
Collapse
Affiliation(s)
- Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Jianing Zhang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chufan Xiao
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Haoying Bai
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
10
|
Prostate Cancer Cell Extracellular Vesicles Increase Mineralisation of Bone Osteoblast Precursor Cells in an In Vitro Model. BIOLOGY 2021; 10:biology10040318. [PMID: 33920233 PMCID: PMC8069461 DOI: 10.3390/biology10040318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/03/2021] [Accepted: 04/07/2021] [Indexed: 12/26/2022]
Abstract
Simple Summary Prostate cancer frequently metastasizes to the bone, where it forms primarily osteoblastic lesions. Currently there is no real therapeutic option for this late stage of disease, and understanding prostate cancer-bone interaction and communication is vital. Using a simple in vitro model of os-teoblast differentiation and mineralization, we studied this interaction and observed that prostate cancer cells secreted large quantities of extracellular vesicles containing microRNAs. When ex-posed to the extracellular vesicles, increased osteoblast differentiation and mineralization could be observed, and upon RNA-seq several of these microRNAs were implicated as upstream regulators of the mineralization process. These microRNAs also correlated with poor survival in online analysis of patient datasets. We characterized and validated four genes known to be targeted by microRNA-16, and found that extracellular vesicles could deliver miR-16, and increase minerali-zation. Abstract Skeletal metastases are the most common form of secondary tumour associated with prostate cancer (PCa). The aberrant function of bone cells neighbouring these tumours leads to the devel-opment of osteoblastic lesions. Communication between PCa cells and bone cells in bone envi-ronments governs both the formation/development of the associated lesion, and growth of the secondary tumour. Using osteoblasts as a model system, we observed that PCa cells and their conditioned medium could stimulate and increase mineralisation and osteoblasts’ differentiation. Secreted factors within PCa-conditioned medium responsible for osteoblastic changes included small extracellular vesicles (sEVs), which were sufficient to drive osteoblastogenesis. Using MiR-seq, we profiled the miRNA content of PCa sEVs, showing that miR-16-5p was highly ex-pressed. MiR-16 was subsequently higher in EV-treated 7F2 cells and a miR-16 mimic could also stimulate mineralisation. Next, using RNA-seq of extracellular vesicle (EV)-treated 7F2 cells, we observed a large degree of gene downregulation and an increased mineralisation. Ingenuity® Pathway Analysis (IPA®) revealed that miR-16-5p (and other miRs) was a likely upstream effec-tor. MiR-16-5p targets in 7F2 cells, possibly involved in osteoblastogenesis, were included for val-idation, namely AXIN2, PLSCR4, ADRB2 and DLL1. We then confirmed the targeting and dow-regulation of these genes by sEV miR-16-5p using luciferase UTR (untranslated region) reporters. Conversely, the overexpression of PLSCR4, ADRB2 and DLL1 lead to decreased osteoblastogene-sis. These results indicate that miR-16 is an inducer of osteoblastogenesis and is transmitted through prostate cancer-derived sEVs. The mechanism is a likely contributor towards the for-mation of osteoblastic lesions in metastatic PCa.
Collapse
|
11
|
Wee NKY, Nguyen AD, Enriquez RF, Zhang L, Herzog H, Baldock PA. Neuropeptide Y Regulation of Energy Partitioning and Bone Mass During Cold Exposure. Calcif Tissue Int 2020; 107:510-523. [PMID: 32804252 DOI: 10.1007/s00223-020-00745-9] [Citation(s) in RCA: 4] [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: 01/13/2020] [Accepted: 08/04/2020] [Indexed: 10/23/2022]
Abstract
The maintenance of whole body energy homeostasis is critical to survival and mechanisms exist whereby an organism can adapt to its environment and the stresses placed upon it. Environmental temperature and thermogenesis are key components known to affect energy balance. However, little is known about how these processes are balanced against the overall energy balance. We show that even mild cold exposure has a significant effect on energy expenditure and UCP-1 levels which increase by 43% and 400%, respectively, when wild-type (WT) mice at thermoneutral (29 °C) were compared to mice at room temperature (22 °C) conditions. Interestingly, bone mass was lower in cold-stressed WT mice with significant reductions in femoral bone mineral content (- 19%) and bone volume (- 13%). Importantly, these cold-induced skeletal changes were absent in mice lacking NPY, one of the main controllers of energy homeostasis, highlighting the critical role of NPY in this process. However, energy expenditure was significantly greater in cold-exposed NPY null mice, indicating that suppression of non-thermogenic tissues, like bone, contributes to the adaptive responses to cold exposure. Altogether, this work identifies NPY as being crucial in coordinating energy and bone homeostasis where it suppresses energy expenditure, UCP-1 levels and lowers bone mass under conditions of cold exposure.
Collapse
Affiliation(s)
- Natalie K Y Wee
- Bone Biology Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Amy D Nguyen
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Ronaldo F Enriquez
- Bone Biology Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Lei Zhang
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
- School of Medical Sciences, University of NSW, Sydney, NSW, Australia
| | - Paul A Baldock
- Bone Biology Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia.
- School of Medical Sciences, University of NSW, Sydney, NSW, Australia.
- School of Medicine Sydney, University of Notre Dame Australia, Sydney, Australia.
| |
Collapse
|
12
|
Abstract
The skeleton is highly vascularized due to the various roles blood vessels play in the homeostasis of bone and marrow. For example, blood vessels provide nutrients, remove metabolic by-products, deliver systemic hormones, and circulate precursor cells to bone and marrow. In addition to these roles, bone blood vessels participate in a variety of other functions. This article provides an overview of the afferent, exchange and efferent vessels in bone and marrow and presents the morphological layout of these blood vessels regarding blood flow dynamics. In addition, this article discusses how bone blood vessels participate in bone development, maintenance, and repair. Further, mechanical loading-induced bone adaptation is presented regarding interstitial fluid flow and pressure, as regulated by the vascular system. The role of the sympathetic nervous system is discussed in relation to blood vessels and bone. Finally, vascular participation in bone accrual with intermittent parathyroid hormone administration, a medication prescribed to combat age-related bone loss, is described and age- and disease-related impairments in blood vessels are discussed in relation to bone and marrow dysfunction. © 2020 American Physiological Society. Compr Physiol 10:1009-1046, 2020.
Collapse
Affiliation(s)
- Rhonda D Prisby
- Bone Vascular and Microcirculation Laboratory, Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
13
|
Abstract
It is from the discovery of leptin and the central nervous system as a regulator of bone remodeling that the presence of autonomic nerves within the skeleton transitioned from a mere histological observation to the mechanism whereby neurons of the central nervous system communicate with cells of the bone microenvironment and regulate bone homeostasis. This shift in paradigm sparked new preclinical and clinical investigations aimed at defining the contribution of sympathetic, parasympathetic, and sensory nerves to the process of bone development, bone mass accrual, bone remodeling, and cancer metastasis. The aim of this article is to review the data that led to the current understanding of the interactions between the autonomic and skeletal systems and to present a critical appraisal of the literature, bringing forth a schema that can put into physiological and clinical context the main genetic and pharmacological observations pointing to the existence of an autonomic control of skeletal homeostasis. The different types of nerves found in the skeleton, their functional interactions with bone cells, their impact on bone development, bone mass accrual and remodeling, and the possible clinical or pathophysiological relevance of these findings are discussed.
Collapse
Affiliation(s)
- Florent Elefteriou
- Department of Molecular and Human Genetics and Orthopedic Surgery, Center for Skeletal Medicine and Biology, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
14
|
Tanaka H, Miura T, Yamashita T, Yoneda M, Takagi S. Characteristics of Bone Strength and Metabolism in Type 2 Diabetic Model Nagoya Shibata Yasuda Mice. Biol Pharm Bull 2018; 41:1567-1573. [PMID: 30012927 DOI: 10.1248/bpb.b18-00275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We evaluated the suitability of Nagoya Shibata Yasuda (NSY) mice as an animal model for examining the influence of a glucose metabolism disorder on bone integrity, using Institute of Cancer Research (ICR) mice as controls. We selected six NSY and ICR mice each that were matched for weight, and measured serum glucose levels, serum insulin levels, and conducted an oral glucose tolerance test. Histological sections of the femurs of both mouse lines were prepared, and the bone strength, mass, and microstructure of the femur were compared, along with bone metabolism. Serum glucose levels were significantly higher in the NSY mice than in the control mice, but body weight and serum insulin levels did not differ between the groups. Bone mass, microstructure, and strength of the femur, and bone metabolism were lower in the NSY mice than in the control mice. In the cortical bone of the femur in the NSY mice, several parts were not stained with eosin, demonstrating a strong negative correlation between serum glucose levels and bone mineral density; however, there was a negative correlation between serum glucose levels and bone metabolic markers. The bone turnover rate in the NSY mice was decreased by hyperglycemia, resulting in a thinner and shorter femur, reduced cortical and trabecular areas, and lower bone mass compared to those of the control mice. Collectively, these results suggest deteriorated bone strength of the femur in NSY mice, serving as a useful model for studying the link between glucose metabolism and bone integrity.
Collapse
Affiliation(s)
- Hiroaki Tanaka
- Graduate School of Health Science, Suzuka University of Medical Science
| | - Toshihiro Miura
- Graduate School of Health Science, Suzuka University of Medical Science
| | - Takenori Yamashita
- Department of Radiological Technology, Faculty of Health Science, Suzuka University of Medical Science
| | - Misao Yoneda
- Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science
| | - Satoshi Takagi
- Department of Physical Therapy, Faculty of Health and Medical Sciences, Tokoha University
| |
Collapse
|
15
|
Robbins A, Tom CATMB, Cosman MN, Moursi C, Shipp L, Spencer TM, Brash T, Devlin MJ. Low temperature decreases bone mass in mice: Implications for humans. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2018; 167:557-568. [PMID: 30187469 DOI: 10.1002/ajpa.23684] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/21/2018] [Accepted: 06/26/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Humans exhibit significant ecogeographic variation in bone size and shape. However, it is unclear how significantly environmental temperature influences cortical and trabecular bone, making it difficult to recognize adaptation versus acclimatization in past populations. There is some evidence that cold-induced bone loss results from sympathetic nervous system activation and can be reduced by nonshivering thermogenesis (NST) via uncoupling protein (UCP1) in brown adipose tissue (BAT). Here we test two hypotheses: (1) low temperature induces impaired cortical and trabecular bone acquisition and (2) UCP1, a marker of NST in BAT, increases in proportion to degree of low-temperature exposure. METHODS We housed wildtype C57BL/6J male mice in pairs at 26 °C (thermoneutrality), 22 °C (standard), and 20 °C (cool) from 3 weeks to 6 or 12 weeks of age with access to food and water ad libitum (N = 8/group). RESULTS Cool housed mice ate more but had lower body fat at 20 °C versus 26 °C. Mice at 20 °C had markedly lower distal femur trabecular bone volume fraction, thickness, and connectivity density and lower midshaft femur cortical bone area fraction versus mice at 26 °C (p < .05 for all). UCP1 expression in BAT was inversely related to temperature. DISCUSSION These results support the hypothesis that low temperature was detrimental to bone mass acquisition. Nonshivering thermogenesis in brown adipose tissue increased in proportion to low-temperature exposure but was insufficient to prevent bone loss. These data show that chronic exposure to low temperature impairs bone architecture, suggesting climate may contribute to phenotypic variation in humans and other hominins.
Collapse
Affiliation(s)
- Amy Robbins
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | | | - Miranda N Cosman
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Cleo Moursi
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Lillian Shipp
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Taylor M Spencer
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Timothy Brash
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Maureen J Devlin
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
16
|
Devlin M, Robbins A, Cosman M, Moursi C, Cloutier A, Louis L, Van Vliet M, Conlon C, Bouxsein M. Differential effects of high fat diet and diet-induced obesity on skeletal acquisition in female C57BL/6J vs. FVB/NJ Mice. Bone Rep 2018; 8:204-214. [PMID: 29955639 PMCID: PMC6020275 DOI: 10.1016/j.bonr.2018.04.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 03/06/2018] [Accepted: 04/16/2018] [Indexed: 12/28/2022] Open
Abstract
The effects of obesity on bone metabolism are complex, and may be mediated by consumption of a high fat diet and/or by obesity-induced metabolic dysregulation. To test the hypothesis that both high fat (HF) diet and diet-induced metabolic disease independently decrease skeletal acquisition, we compared effects of HF diet on bone mass and microarchitecture in two mouse strains: diet-induced obesity (DIO)-susceptible C57BL/6J (B6) and DIO-resistant FVB/NJ (FVB). At 3 wks of age we weaned 120 female FVB and B6 mice onto normal (N, 10% Kcal/fat) or HF diet (45% Kcal/fat) and euthanized them at 6, 12 and 20 weeks of age (N = 10/grp). Outcomes included body mass; percent fat and whole-body bone mineral density (WBBMD, g/cm2) via DXA; cortical and trabecular bone architecture at the midshaft and distal femur via μCT; and marrow adiposity via histomorphometry. In FVB HF, body mass, percent body fat, WBBMD and marrow adiposity did not differ vs. N, but trabecular bone mass was lower at 6 wks of age only (p < 0.05), cortical bone geometric properties were lower at 12 wks only, and bone strength was lower at 20 wks of age only in HF vs. N (p < 0.05). In contrast, B6 HF had higher body mass, percent body fat, and leptin vs. N. B6 HF also had higher WBBMD (p < 0.05) at 9 and 12 wks of age but lower distal femur trabecular bone mass at 12 wks of age, and lower body mass-adjusted cortical bone properties at 20 wks of age compared to N (p < 0.05). Marrow adiposity was also markedly higher in B6 HF vs. N. Overall, HF diet negatively affected bone mass in both strains, but was more deleterious to trabecular bone microarchitecture and marrow adiposity in B6 than in FVB mice. These data suggest that in addition to fat consumption itself, the metabolic response to high fat diet independently alters skeletal acquisition in obesity.
Collapse
Affiliation(s)
- M.J. Devlin
- Department of Anthropology, University of Michigan, Ann Arbor, MI 48104, United States
| | - A. Robbins
- Department of Anthropology, University of Michigan, Ann Arbor, MI 48104, United States
| | - M.N. Cosman
- Department of Anthropology, University of Michigan, Ann Arbor, MI 48104, United States
| | - C.A. Moursi
- Department of Anthropology, University of Michigan, Ann Arbor, MI 48104, United States
| | - A.M. Cloutier
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, United States
| | - L. Louis
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, United States
| | - M. Van Vliet
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, United States
| | - C. Conlon
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, United States
| | - M.L. Bouxsein
- Harvard Medical School, Boston, MA 02215, United States
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, United States
| |
Collapse
|
17
|
Goetz TG, Mamillapalli R, Devlin MJ, Robbins AE, Majidi-Zolbin M, Taylor HS. Cross-sex testosterone therapy in ovariectomized mice: addition of low-dose estrogen preserves bone architecture. Am J Physiol Endocrinol Metab 2017; 313:E540-E551. [PMID: 28765273 PMCID: PMC5792142 DOI: 10.1152/ajpendo.00161.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 01/17/2023]
Abstract
Cross-sex hormone therapy (XHT) is widely used by transgender people to alter secondary sex characteristics to match their desired gender presentation. Here, we investigate the long-term effects of XHT on bone health using a murine model. Female mice underwent ovariectomy at either 6 or 10 wk and began weekly testosterone or vehicle injections. Dual-energy X-ray absorptiometry (DXA) was performed (20 wk) to measure bone mineral density (BMD), and microcomputed tomography was performed to compare femoral cortical and trabecular bone architecture. The 6-wk testosterone group had comparable BMD with controls by DXA but reduced bone volume fraction, trabecular number, and cortical area fraction and increased trabecular separation by microcomputed tomography. Ten-week ovariectomy/XHT maintained microarchitecture, suggesting that estrogen is critical for bone acquisition during adolescence and that late, but not early, estrogen loss can be sufficiently replaced by testosterone alone. Given these findings, we then compared effects of testosterone with effects of weekly estrogen or combined testosterone/low-dose estrogen treatment after a 6-wk ovariectomy. Estrogen treatment increased spine BMD and microarchitecture, including bone volume fraction, trabecular number, trabecular thickness, and connectivity density, and decreased trabecular separation. Combined testosterone-estrogen therapy caused similar increases in femur and spine BMD and improved architecture (increased bone volume fraction, trabecular number, trabecular thickness, and connectivity density) to estrogen therapy and were superior compared with mice treated with testosterone only. These results demonstrate estradiol is critical for bone acquisition and suggest a new cross-sex hormone therapy adding estrogens to testosterone treatments with potential future clinical implications for treating transgender youth or men with estrogen deficiency.
Collapse
Affiliation(s)
- Teddy G Goetz
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut; and
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut; and
| | - Maureen J Devlin
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Amy E Robbins
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan
| | - Masoumeh Majidi-Zolbin
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut; and
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut; and
| |
Collapse
|
18
|
Isoproterenol Increases RANKL Expression in a ATF4/NFATc1-Dependent Manner in Mouse Osteoblastic Cells. Int J Mol Sci 2017; 18:ijms18102204. [PMID: 29053621 PMCID: PMC5666884 DOI: 10.3390/ijms18102204] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/16/2017] [Accepted: 10/20/2017] [Indexed: 12/12/2022] Open
Abstract
Sympathetic nervous system stimulation-induced β-adrenergic signal transduction is known to induce bone loss and increase of osteoclast activity. Although isoproterenol, a nonspecific β-adrenergic receptor agonist, has been shown to increase receptor activator of NF-κB ligand (RANKL), the details of the regulatory mechanisms remain unclear. In the present study, we investigated the role of the nuclear factor of activated T-cells (NFAT) in isoproterenol-induced RANKL expression in C2C12 and in primary cultured mouse calvarial cells. Isoproterenol increased nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) and RANKL expressions at both mRNA and protein levels and increased NFAT reporter activity. NFATc1 knockdown blocked isoproterenol-mediated RANKL expression. Isoproterenol also promoted cAMP response element-binding protein 1 (CREB1) and activating transcription factor 4 (ATF4) phosphorylation. Isoproterenol-mediated transcriptional activation of NFAT was blocked by protein kinase A (PKA) inhibitor H89. Isoproterenol-induced CREB1, ATF4, NFATc1, and RANKL expressions were suppressed by H89. Mutations in cAMP response element-like or NFAT-binding element suppressed isoproterenol-induced RANKL promoter activity. Chromatin immunoprecipitation analysis demonstrated that isoproterenol increased NFAT-binding and ATF4-binding activities on the mouse RANKL promoter, but did not increase CREB1-binding activity. Association of NFATc1 and ATF4 was not observed in a co-immunoprecipitation study. ATF4 knockdown suppressed isoproterenol-induced NFAT binding to the RANKL promoter, whereas NFATc1 knockdown did not suppress isoproterenol-induced ATF4 binding to the RANKL promoter. ATF4 knockdown suppressed isoproterenol-induced expressions of NFATc1 and RANKL. These results suggest that isoproterenol increases RANKL expression in an ATF4/NFATc1-dependent manner.
Collapse
|
19
|
Craft CS, Scheller EL. Evolution of the Marrow Adipose Tissue Microenvironment. Calcif Tissue Int 2017; 100:461-475. [PMID: 27364342 PMCID: PMC5618436 DOI: 10.1007/s00223-016-0168-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/21/2016] [Indexed: 12/29/2022]
Abstract
Adipocytes of the marrow adipose tissue (MAT) are distributed throughout the skeleton, are embedded in extracellular matrix, and are surrounded by cells of the hematopoietic and osteogenic lineages. MAT is a persistent component of the skeletal microenvironment and has the potential to impact local processes including bone accrual and hematopoietic function. In this review, we discuss the initial evolution of MAT in vertebrate lineages while emphasizing comparisons to the development of peripheral adipose, hematopoietic, and skeletal tissues. We then apply these evolutionary clues to define putative functions of MAT. Lastly, we explore the regulation of MAT by two major components of its microenvironment, the extracellular matrix and the nerves embedded within. The extracellular matrix and nerves contribute to both rapid and continuous modification of the MAT niche and may help to explain evolutionary conserved mechanisms underlying the coordinated regulation of blood, bone, and MAT within the skeleton.
Collapse
Affiliation(s)
- Clarissa S Craft
- Department of Cell Biology & Physiology, Washington University, Saint Louis, MO, 63110, USA
- Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University, Saint Louis, MO, 63110, USA
| | - Erica L Scheller
- Department of Cell Biology & Physiology, Washington University, Saint Louis, MO, 63110, USA.
- Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University, Saint Louis, MO, 63110, USA.
| |
Collapse
|
20
|
Chen D, Wang Z. Adrenaline inhibits osteogenesis via repressing miR-21 expression. Cell Biol Int 2016; 41:8-15. [DOI: 10.1002/cbin.10685] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/17/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Danying Chen
- Department of Dental Implantology, School and Hospital of Stomatology; Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration; 399 Yanchang Road Shanghai 200072 PR China
| | - Zuolin Wang
- Department of Dental Implantology, School and Hospital of Stomatology; Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration; 399 Yanchang Road Shanghai 200072 PR China
| |
Collapse
|
21
|
Yao Q, Liang H, Huang B, Xiang L, Wang T, Xiong Y, Yang B, Guo Y, Gong P. Beta-adrenergic signaling affect osteoclastogenesis via osteocytic MLO-Y4 cells' RANKL production. Biochem Biophys Res Commun 2016; 488:634-640. [PMID: 27823934 DOI: 10.1016/j.bbrc.2016.11.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/03/2016] [Indexed: 02/05/2023]
Abstract
The sympathetic nervous system play a pivotal role in bone remodeling through β-adrenoceptor (β-AR). However, it is not well documented whether the β-adrenoceptor pathway has the potential to influence osteocytes. In this study, cell viability, the expression of β-AR subtypes, enzymes of catecholamine synthesis or degradation, bone-related gene and protein in osteocytic MLO-Y4 cells were investigated by β-adrenergic stimulation. Isoproterenol (ISO) promoted RANKL to OPG expression in osteocytes, as well as osteoclasts formation in osteocytes-RAW264.7 cell co-cultures but not RAW264.7 cell monoculture. The ISO-stimulated effect was enhanced in β1-AR antagonist pretreatment, but was rescued by blocking β2-AR. The results indicate that β1-and β2-AR play reciprocal roles on MLO-Y4 cells in the regulation of osteoclastogenesis, and osteocyte β-adrenergic signaling might be a new valuable therapy for bone disease.
Collapse
Affiliation(s)
- Qianqian Yao
- Oral Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hengxing Liang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bo Huang
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, China; State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Xiang
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, China; State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tianlu Wang
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, China; State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Xiong
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, China; State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Yang
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, China; State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanjun Guo
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, China; State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, China; State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
22
|
Laurent MR, Dubois V, Claessens F, Verschueren SMP, Vanderschueren D, Gielen E, Jardí F. Muscle-bone interactions: From experimental models to the clinic? A critical update. Mol Cell Endocrinol 2016; 432:14-36. [PMID: 26506009 DOI: 10.1016/j.mce.2015.10.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/13/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023]
Abstract
Bone is a biomechanical tissue shaped by forces from muscles and gravitation. Simultaneous bone and muscle decay and dysfunction (osteosarcopenia or sarco-osteoporosis) is seen in ageing, numerous clinical situations including after stroke or paralysis, in neuromuscular dystrophies, glucocorticoid excess, or in association with vitamin D, growth hormone/insulin like growth factor or sex steroid deficiency, as well as in spaceflight. Physical exercise may be beneficial in these situations, but further work is still needed to translate acceptable and effective biomechanical interventions like vibration therapy from animal models to humans. Novel antiresorptive and anabolic therapies are emerging for osteoporosis as well as drugs for sarcopenia, cancer cachexia or muscle wasting disorders, including antibodies against myostatin or activin receptor type IIA and IIB (e.g. bimagrumab). Ideally, increasing muscle mass would increase muscle strength and restore bone loss from disuse. However, the classical view that muscle is unidirectionally dominant over bone via mechanical loading is overly simplistic. Indeed, recent studies indicate a role for neuronal regulation of not only muscle but also bone metabolism, bone signaling pathways like receptor activator of nuclear factor kappa-B ligand (RANKL) implicated in muscle biology, myokines affecting bone and possible bone-to-muscle communication. Moreover, pharmacological strategies inducing isolated myocyte hypertrophy may not translate into increased muscle power because tendons, connective tissue, neurons and energy metabolism need to adapt as well. We aim here to critically review key musculoskeletal molecular pathways involved in mechanoregulation and their effect on the bone-muscle unit as a whole, as well as preclinical and emerging clinical evidence regarding the effects of sarcopenia therapies on osteoporosis and vice versa.
Collapse
Affiliation(s)
- Michaël R Laurent
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium.
| | - Vanessa Dubois
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Frank Claessens
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Sabine M P Verschueren
- Research Group for Musculoskeletal Rehabilitation, Department of Rehabilitation Science, KU Leuven, 3000 Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Evelien Gielen
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ferran Jardí
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
23
|
Devlin MJ, Brooks DJ, Conlon C, Vliet MV, Louis L, Rosen CJ, Bouxsein ML. Daily leptin blunts marrow fat but does not impact bone mass in calorie-restricted mice. J Endocrinol 2016; 229:295-306. [PMID: 27340200 PMCID: PMC5171226 DOI: 10.1530/joe-15-0473] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 04/11/2016] [Indexed: 01/02/2023]
Abstract
Starvation induces low bone mass and high bone marrow adiposity in humans, but the underlying mechanisms are poorly understood. The adipokine leptin falls in starvation, suggesting that hypoleptinemia may be a link between negative energy balance, bone marrow fat accumulation, and impaired skeletal acquisition. In that case, treating mice with leptin during caloric restriction (CR) should reduce marrow adipose tissue (MAT) and improve bone mass. To test this hypothesis, female C57Bl/6J mice were fed a 30% CR or normal (N) diet from 5 to 10 weeks of age, with daily injections of vehicle (VEH), 1mg/kg leptin (LEP1), or 2mg/kg leptin (LEP2) (N=6-8/group). Outcomes included body mass, body fat percentage, and whole-body bone mineral density (BMD) via peripheral dual-energy X-ray absorptiometry, cortical and trabecular microarchitecture via microcomputed tomography (μCT), and MAT volume via μCT of osmium tetroxide-stained bones. Overall, CR mice had lower body mass, body fat percentage, BMD, and cortical bone area fraction, but more connected trabeculae, vs N mice (P<0.05 for all). Most significantly, although MAT was elevated in CR vs N overall, leptin treatment blunted MAT formation in CR mice by 50% vs VEH (P<0.05 for both leptin doses). CR LEP2 mice weighed less vs CR VEH mice at 9-10 weeks of age (P<0.05), but leptin treatment did not affect body fat percentage, BMD, or bone microarchitecture within either diet. These data demonstrate that once daily leptin bolus during CR inhibits bone marrow adipose expansion without affecting bone mass acquisition, suggesting that leptin has distinct effects on starvation-induced bone marrow fat formation and skeletal acquisition.
Collapse
Affiliation(s)
- M J Devlin
- Department of AnthropologyUniversity of Michigan, Ann Arbor, Michigan, USA
| | - D J Brooks
- Center for Advanced Orthopedic StudiesBeth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - C Conlon
- Center for Advanced Orthopedic StudiesBeth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - M van Vliet
- Center for Advanced Orthopedic StudiesBeth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - L Louis
- Center for Advanced Orthopedic StudiesBeth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - C J Rosen
- Maine Medical Center Research InstituteScarborough, Maine, USA
| | - M L Bouxsein
- Center for Advanced Orthopedic StudiesBeth Israel Deaconess Medical Center, Boston, Massachusetts, USA Harvard Medical SchoolBoston, Massachusetts, USA
| |
Collapse
|
24
|
Tascau L, Gardner T, Anan H, Yongpravat C, Cardozo CP, Bauman WA, Lee FY, Oh DS, Tawfeek HA. Activation of Protein Kinase A in Mature Osteoblasts Promotes a Major Bone Anabolic Response. Endocrinology 2016; 157:112-26. [PMID: 26488807 DOI: 10.1210/en.2015-1614] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Protein kinase A (PKA) regulates osteoblast cell function in vitro and is activated by important bone mass modulating agents. We determined whether PKA activation in osteoblasts is sufficient to mediate a bone anabolic response. Thus, a mouse model conditionally expressing a constitutively active PKA (CA-PKA) in osteoblasts (CA-PKA-OB mouse) was developed by crossing a 2.3-kb α1 (I)-collagen promoter-Cre mouse with a floxed-CA-PKA mouse. Primary osteoblasts from the CA-PKA-OB mice exhibited higher basal PKA activity than those from control mice. Microcomputed tomographic analysis revealed that CA-PKA-OB female mice had an 8.6-fold increase in femoral but only 1.16-fold increase in lumbar 5 vertebral bone volume/total volume. Femur cortical thickness and volume were also higher in the CA-PKA-OB mice. In contrast, alterations in many femoral microcomputed tomographic parameters in male CA-PKA-OB mice were modest. Interestingly, the 3-dimensional structure model index was substantially lower both in femur and lumbar 5 of male and female CA-PKA-OB mice, reflecting an increase in the plate to rod-like structure ratio. In agreement, femurs from female CA-PKA-OB mice had greater load to failure and were stiffer compared with those of control mice. Furthermore, the CA-PKA-OB mice had higher levels of serum bone turnover markers and increased osteoblast and osteoclast numbers per total tissue area compared with control animals. In summary, constitutive activation of PKA in osteoblasts is sufficient to increase bone mass and favorably modify bone architecture and improve mechanical properties. PKA activation in mature osteoblasts is, therefore, an important target for designing anabolic drugs for treating diseases with bone loss.
Collapse
Affiliation(s)
- Liana Tascau
- National Center for the Medical Consequences of Spinal Cord Injury (C.P.C., W.A.B., H.A.T.), James J. Peters VA Medical Center, Bronx, New York 10468; Center for Orthopaedic Research (T.G., C.Y., F.Y.L.), College of Dental Medicine (D.S.O.), and Department of Molecular Medicine (L.T.), Columbia University, and Departments of Medicine (C.P.C., W.A.B., H.A.T.), Rehabilitation Medicine (C.P.C., W.A.B.), and Pharmacology and Systems Therapeutics (C.P.C.), The Icahn School of Medicine at Mount Sinai, New York, New York 10029; and Sacred Heart Hospital/Temple University (H.A.), Allentown, Pennsylvania 16102
| | - Thomas Gardner
- National Center for the Medical Consequences of Spinal Cord Injury (C.P.C., W.A.B., H.A.T.), James J. Peters VA Medical Center, Bronx, New York 10468; Center for Orthopaedic Research (T.G., C.Y., F.Y.L.), College of Dental Medicine (D.S.O.), and Department of Molecular Medicine (L.T.), Columbia University, and Departments of Medicine (C.P.C., W.A.B., H.A.T.), Rehabilitation Medicine (C.P.C., W.A.B.), and Pharmacology and Systems Therapeutics (C.P.C.), The Icahn School of Medicine at Mount Sinai, New York, New York 10029; and Sacred Heart Hospital/Temple University (H.A.), Allentown, Pennsylvania 16102
| | - Hussein Anan
- National Center for the Medical Consequences of Spinal Cord Injury (C.P.C., W.A.B., H.A.T.), James J. Peters VA Medical Center, Bronx, New York 10468; Center for Orthopaedic Research (T.G., C.Y., F.Y.L.), College of Dental Medicine (D.S.O.), and Department of Molecular Medicine (L.T.), Columbia University, and Departments of Medicine (C.P.C., W.A.B., H.A.T.), Rehabilitation Medicine (C.P.C., W.A.B.), and Pharmacology and Systems Therapeutics (C.P.C.), The Icahn School of Medicine at Mount Sinai, New York, New York 10029; and Sacred Heart Hospital/Temple University (H.A.), Allentown, Pennsylvania 16102
| | - Charlie Yongpravat
- National Center for the Medical Consequences of Spinal Cord Injury (C.P.C., W.A.B., H.A.T.), James J. Peters VA Medical Center, Bronx, New York 10468; Center for Orthopaedic Research (T.G., C.Y., F.Y.L.), College of Dental Medicine (D.S.O.), and Department of Molecular Medicine (L.T.), Columbia University, and Departments of Medicine (C.P.C., W.A.B., H.A.T.), Rehabilitation Medicine (C.P.C., W.A.B.), and Pharmacology and Systems Therapeutics (C.P.C.), The Icahn School of Medicine at Mount Sinai, New York, New York 10029; and Sacred Heart Hospital/Temple University (H.A.), Allentown, Pennsylvania 16102
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury (C.P.C., W.A.B., H.A.T.), James J. Peters VA Medical Center, Bronx, New York 10468; Center for Orthopaedic Research (T.G., C.Y., F.Y.L.), College of Dental Medicine (D.S.O.), and Department of Molecular Medicine (L.T.), Columbia University, and Departments of Medicine (C.P.C., W.A.B., H.A.T.), Rehabilitation Medicine (C.P.C., W.A.B.), and Pharmacology and Systems Therapeutics (C.P.C.), The Icahn School of Medicine at Mount Sinai, New York, New York 10029; and Sacred Heart Hospital/Temple University (H.A.), Allentown, Pennsylvania 16102
| | - William A Bauman
- National Center for the Medical Consequences of Spinal Cord Injury (C.P.C., W.A.B., H.A.T.), James J. Peters VA Medical Center, Bronx, New York 10468; Center for Orthopaedic Research (T.G., C.Y., F.Y.L.), College of Dental Medicine (D.S.O.), and Department of Molecular Medicine (L.T.), Columbia University, and Departments of Medicine (C.P.C., W.A.B., H.A.T.), Rehabilitation Medicine (C.P.C., W.A.B.), and Pharmacology and Systems Therapeutics (C.P.C.), The Icahn School of Medicine at Mount Sinai, New York, New York 10029; and Sacred Heart Hospital/Temple University (H.A.), Allentown, Pennsylvania 16102
| | - Francis Y Lee
- National Center for the Medical Consequences of Spinal Cord Injury (C.P.C., W.A.B., H.A.T.), James J. Peters VA Medical Center, Bronx, New York 10468; Center for Orthopaedic Research (T.G., C.Y., F.Y.L.), College of Dental Medicine (D.S.O.), and Department of Molecular Medicine (L.T.), Columbia University, and Departments of Medicine (C.P.C., W.A.B., H.A.T.), Rehabilitation Medicine (C.P.C., W.A.B.), and Pharmacology and Systems Therapeutics (C.P.C.), The Icahn School of Medicine at Mount Sinai, New York, New York 10029; and Sacred Heart Hospital/Temple University (H.A.), Allentown, Pennsylvania 16102
| | - Daniel S Oh
- National Center for the Medical Consequences of Spinal Cord Injury (C.P.C., W.A.B., H.A.T.), James J. Peters VA Medical Center, Bronx, New York 10468; Center for Orthopaedic Research (T.G., C.Y., F.Y.L.), College of Dental Medicine (D.S.O.), and Department of Molecular Medicine (L.T.), Columbia University, and Departments of Medicine (C.P.C., W.A.B., H.A.T.), Rehabilitation Medicine (C.P.C., W.A.B.), and Pharmacology and Systems Therapeutics (C.P.C.), The Icahn School of Medicine at Mount Sinai, New York, New York 10029; and Sacred Heart Hospital/Temple University (H.A.), Allentown, Pennsylvania 16102
| | - Hesham A Tawfeek
- National Center for the Medical Consequences of Spinal Cord Injury (C.P.C., W.A.B., H.A.T.), James J. Peters VA Medical Center, Bronx, New York 10468; Center for Orthopaedic Research (T.G., C.Y., F.Y.L.), College of Dental Medicine (D.S.O.), and Department of Molecular Medicine (L.T.), Columbia University, and Departments of Medicine (C.P.C., W.A.B., H.A.T.), Rehabilitation Medicine (C.P.C., W.A.B.), and Pharmacology and Systems Therapeutics (C.P.C.), The Icahn School of Medicine at Mount Sinai, New York, New York 10029; and Sacred Heart Hospital/Temple University (H.A.), Allentown, Pennsylvania 16102
| |
Collapse
|
25
|
Poon CCW, Li RWS, Seto SW, Kong SK, Ho HP, Hoi MPM, Lee SMY, Ngai SM, Chan SW, Leung GPH, Kwan YW. In vitro vitamin K(2) and 1α,25-dihydroxyvitamin D(3) combination enhances osteoblasts anabolism of diabetic mice. Eur J Pharmacol 2015; 767:30-40. [PMID: 26452518 DOI: 10.1016/j.ejphar.2015.09.048] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/25/2015] [Accepted: 09/29/2015] [Indexed: 12/20/2022]
Abstract
In this study, we evaluated the anabolic effect and the underlying cellular mechanisms involved of vitamin K2 (10 nM) and 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) (10 nM), alone and in combination, on primary osteoblasts harvested from the iliac crests of C57BL/KsJ lean (+/+) and obese/diabetic (db/db) mice. A lower alkaline phosphatase (ALP) activity plus a reduced expression of bone anabolic markers and bone formation transcription factors (osteocalcin, Runx2, Dlx5, ATF4 and OSX) were consistently detected in osteoblasts of db/db mice compared to lean mice. A significantly higher calcium deposits formation in osteoblasts was observed in lean mice when compared to db/db mice. Co-administration of vitamin K2 (10 nM) and 1,25(OH)2D3 (10 nM) caused an enhancement of calcium deposits in osteoblasts in both strains of mice. Vitamins K2 and 1,25(OH)2D3 co-administration time-dependently (7, 14 and 21 days) increased the levels of bone anabolic markers and bone formation transcription factors, with a greater magnitude of increase observed in osteoblasts of db/db mice. Combined vitamins K2 plus 1,25(OH)2D3 treatment significantly enhanced migration and the re-appearance of surface microvilli and ruffles of osteoblasts of db/db mice. Thus, our results illustrate that vitamins K2 plus D3 combination could be a novel therapeutic strategy in treating diabetes-associated osteoporosis.
Collapse
Affiliation(s)
- Christina C W Poon
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Rachel W S Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; Department of Pharmacology and Pharmacy, Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Sai Wang Seto
- National Institute of Complementary Medicine, School of Science and Health, University of Western Sydney, Locked Bag 1797, Penrith, NSW 2751, Australia
| | - Siu Kai Kong
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong
| | - Ho Pui Ho
- Department of Electronic Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong
| | - Maggie P M Hoi
- Institute of Chinese Medical Sciences, The University of Macau, Macau, China
| | - Simon M Y Lee
- Institute of Chinese Medical Sciences, The University of Macau, Macau, China
| | - Sai Ming Ngai
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong
| | - Shun Wan Chan
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong
| | - George P H Leung
- Department of Pharmacology and Pharmacy, Faculty of Medicine, The University of Hong Kong, Hong Kong.
| | - Yiu Wa Kwan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
26
|
Brommage R, Liu J, Doree D, Yu W, Powell DR, Melissa Yang Q. Adult Tph2 knockout mice without brain serotonin have moderately elevated spine trabecular bone but moderately low cortical bone thickness. BONEKEY REPORTS 2015; 4:718. [PMID: 26229596 DOI: 10.1038/bonekey.2015.87] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 04/16/2015] [Indexed: 12/19/2022]
Abstract
Disruption of serotonin synthesis in neurons and the periphery by knockout (KO) of mouse genes for tryptophan hydroxylases (peripheral Tph1 and neuronal Tph2) has been claimed to decrease (Tph2 KO) and increase (Tph1 KO) bone mass. In this report, adult male and female Tph2 KO mice were observed to have elevated spine trabecular bone. Female Tph2 KO mice have reduced midshaft femur cortical bone thickness. Bone mass was normal in male and female Tph1 KO mice examined as part of a Tph1/Tph2 double knockout (DKO) mouse cohort.
Collapse
Affiliation(s)
| | - Jeff Liu
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| | - Deon Doree
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| | - Wangsheng Yu
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| | | | | |
Collapse
|
27
|
Veldhuis-Vlug AG, Oei L, Souverein PC, Tanck MWT, Rivadeneira F, Zillikens MC, Kamphuisen PW, Maitland - van der Zee A, de Groot MCH, Hofman A, Uitterlinden AG, Fliers E, de Boer A, Bisschop PH. Association of polymorphisms in the beta-2 adrenergic receptor gene with fracture risk and bone mineral density. Osteoporos Int 2015; 26:2019-27. [PMID: 25910744 PMCID: PMC4483183 DOI: 10.1007/s00198-015-3087-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 02/20/2015] [Indexed: 12/18/2022]
Abstract
UNLABELLED Signaling through the beta-2 adrenergic receptor (B2AR) on the osteoblast influences bone remodeling in rodents. In the B2AR gene, three polymorphisms influence receptor function. We show that these polymorphisms are not associated with fracture risk or bone mineral density in the UCP, Rotterdam Study, and GEFOS cohorts. INTRODUCTION Signaling through the beta-2 adrenergic receptor (B2AR) on the osteoblast influences bone remodeling in rodents. In the B2AR gene, three polymorphisms are known to influence receptor function in vitro and in vivo (rs1042713, rs1042714, and rs1800888). We examined the role of these polymorphisms in the B2AR gene on human bone metabolism. METHODS We performed nested case-control studies to determine the association of these polymorphisms with fracture risk in the Utrecht Cardiovascular Pharmacogenetics (UCP) cohort and in three cohorts of the Rotterdam Study. We also determined the association of these polymorphisms with bone mineral density (BMD) in the GEFOS Consortium. UCP contains drug-dispensing histories from community pharmacies linked to national registrations of hospital discharges in the Netherlands. The Rotterdam Study is a prospective cohort study investigating demographics and risk factors of chronic diseases. GEFOS is a large international collaboration studying the genetics of osteoporosis. Fractures were defined by ICD-9 codes 800-829 in the UCP cohort (158 cases and 2617 unmatched controls) and by regular X-ray examinations, general practitioner, and hospital records in the Rotterdam Study (2209 cases and 8559 unmatched controls). BMD was measured at the femoral neck and lumbar spine using dual-energy X-ray absorptiometry in GEFOS (N = 32,961). RESULTS Meta-analysis of the two nested case-control studies showed pooled odds ratios of 0.98 (0.91-1.05, p = 0.52), 1.04 (0.97-1.12, p = 0.28), and 1.16 (0.83-1.62, p = 0.38) for the associations between rs1042713, rs1042714, and rs1800888 per minor allele and fractures, respectively. There were no significant associations of the polymorphisms and BMD in GEFOS. CONCLUSION In conclusion, polymorphisms in the beta-2 adrenergic receptor gene are not associated with fracture risk or BMD.
Collapse
Affiliation(s)
- A. G. Veldhuis-Vlug
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD Amsterdam, The Netherlands
| | - L. Oei
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium of Healthy Aging (NCHA), Leiden, The Netherlands
| | - P. C. Souverein
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - M. W. T. Tanck
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - F. Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium of Healthy Aging (NCHA), Leiden, The Netherlands
| | - M. C. Zillikens
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium of Healthy Aging (NCHA), Leiden, The Netherlands
| | - P. W. Kamphuisen
- Department of Vascular Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A.H. Maitland - van der Zee
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - M. C. H. de Groot
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - A. Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium of Healthy Aging (NCHA), Leiden, The Netherlands
| | - A. G. Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium of Healthy Aging (NCHA), Leiden, The Netherlands
| | - E. Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD Amsterdam, The Netherlands
| | - A. de Boer
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - P. H. Bisschop
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD Amsterdam, The Netherlands
| |
Collapse
|
28
|
|
29
|
Komori T. Animal models for osteoporosis. Eur J Pharmacol 2015; 759:287-94. [PMID: 25814262 DOI: 10.1016/j.ejphar.2015.03.028] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/08/2015] [Accepted: 03/12/2015] [Indexed: 11/30/2022]
Abstract
The major types of osteoporosis in humans are postmenopausal osteoporosis, disuse osteoporosis, and glucocorticoid-induced osteoporosis. Animal models for postmenopausal osteoporosis are generated by ovariectomy. Bone loss occurs in estrogen deficiency due to enhanced bone resorption and impaired osteoblast function. Estrogen receptor α induces osteoclast apoptosis, but the mechanism for impaired osteoblast function remains to be clarified. Animal models for unloading are generated by tail suspension or hind limb immobilization by sciatic neurectomy, tenotomy, or using plaster cast. Unloading inhibits bone formation and enhances bone resorption, and the involvement of the sympathetic nervous system in it needs to be further investigated. The osteocyte network regulates bone mass by responding to mechanical stress. Osteoblast-specific BCL2 transgenic mice, in which the osteocyte network is completely disrupted, can be a mouse model for the evaluation of osteocyte functions. Glucocorticoid treatment inhibits bone formation and enhances bone resorption, and markedly reduces cancellous bone in humans and large animals, but not consistently in rodents.
Collapse
Affiliation(s)
- Toshihisa Komori
- Department of Cell Biology, Unit of Basic Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan.
| |
Collapse
|
30
|
Abstract
Bone metabolism is regulated by the action of two skeletal cells: osteoblasts and osteoclasts. This process is controlled by many genetic, hormonal and lifestyle factors, but today more and more studies have allowed us to identify a neuronal regulation system termed 'bone-brain crosstalk', which highlights a direct relationship between bone tissue and the nervous system. The first documentation of an anatomic relationship between nerves and bone was made via a wood cut by Charles Estienne in Paris in 1545. His diagram demonstrated nerves entering and leaving the bones of a skeleton. Later, several studies were conducted on bone innervation and, as of today, many observations on the regulation of bone remodeling by neurons and neuropeptides that reside in the CNS have created a new research field, that is, neuroskeletal research.
Collapse
Affiliation(s)
- Alessia Metozzi
- a 1 Department of Surgery and Translational Medicine, Metabolic Bone Diseases Unit, University of Florence, Largo Palagi 1, 50138 Florence, Italy
| | - Lorenzo Bonamassa
- a 1 Department of Surgery and Translational Medicine, Metabolic Bone Diseases Unit, University of Florence, Largo Palagi 1, 50138 Florence, Italy
| | - Gemma Brandi
- b 2 Public Mental Health system 1-4 of Florence, Florence, Italy
| | - Maria Luisa Brandi
- c 3 Department of Surgery and Translational Medicine, Metabolic Bone Diseases Unit, AOUC Careggi, University of Florence, Largo Palagi 1, 50138 Florence, Italy
| |
Collapse
|
31
|
Veldhuis-Vlug AG, Tanck MW, Limonard EJ, Endert E, Heijboer AC, Lips P, Fliers E, Bisschop PH. The effects of beta-2 adrenergic agonist and antagonist on human bone metabolism: a randomized controlled trial. Bone 2015; 71:196-200. [PMID: 25451321 DOI: 10.1016/j.bone.2014.10.024] [Citation(s) in RCA: 19] [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: 07/07/2014] [Revised: 10/15/2014] [Accepted: 10/20/2014] [Indexed: 11/20/2022]
Abstract
PURPOSE Genetic knockout or pharmacological inhibition of the beta-2 adrenergic receptor (B2AR) increased bone mass, whereas stimulation decreased bone mass in rodents. In humans, observational studies support sympathetic nervous system regulation of bone metabolism, but intervention studies are lacking. We aimed to determine the effects of a selective beta-2 adrenergic agonist and non-selective antagonist on human bone metabolism. METHODS 32 healthy postmenopausal women were included in a randomized controlled trial conducted in the Academic Medical Center Amsterdam. Participants were randomized to receive treatment with 17-β estradiol 2mg/day; 17-β estradiol 2mg/day and terbutaline 5mg/day (selective B2AR agonist); propranolol 80mg/day (non-selective B-AR antagonist); or no treatment during 12weeks. Main outcome measure was the change in serum concentrations of procollagen type I N propeptide (P1NP) and C-terminal crosslinking telopeptides of collagen type I (CTx) as markers of bone formation and resorption after 12weeks compared between the treatment groups. Data were analyzed with mixed model analysis. RESULTS 17-β estradiol decreased bone turnover compared to control (P1NP p<0.001, CTx p=0.003), but terbutaline combined with 17-β estradiol failed to increase bone turnover compared to 17-β estradiol alone (P1NP p=0.135, CTx p=0.406). Propranolol did not affect bone turnover compared to control (P1NP p=0.709, CTx p=0.981). CONCLUSION Selective beta-2 adrenergic agonists and non-selective beta-antagonists do not affect human bone turnover although we cannot exclude small changes below the detection limit of this study.
Collapse
Affiliation(s)
- A G Veldhuis-Vlug
- Dept. of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD Amsterdam, The Netherlands.
| | - M W Tanck
- Dept. of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD Amsterdam, The Netherlands.
| | - E J Limonard
- Dept. of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD Amsterdam, The Netherlands.
| | - E Endert
- Dept. of Clinical Chemistry, Laboratory of Endocrinology, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD Amsterdam, The Netherlands.
| | - A C Heijboer
- Dept. of Clinical Chemistry, Endocrine Laboratory, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands.
| | - P Lips
- Dept. of Internal Medicine, Endocrine Section, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands.
| | - E Fliers
- Dept. of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD Amsterdam, The Netherlands.
| | - P H Bisschop
- Dept. of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD Amsterdam, The Netherlands.
| |
Collapse
|
32
|
Vanderschueren D, Laurent MR, Claessens F, Gielen E, Lagerquist MK, Vandenput L, Börjesson AE, Ohlsson C. Sex steroid actions in male bone. Endocr Rev 2014; 35:906-60. [PMID: 25202834 PMCID: PMC4234776 DOI: 10.1210/er.2014-1024] [Citation(s) in RCA: 190] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sex steroids are chief regulators of gender differences in the skeleton, and male gender is one of the strongest protective factors against osteoporotic fractures. This advantage in bone strength relies mainly on greater cortical bone expansion during pubertal peak bone mass acquisition and superior skeletal maintenance during aging. During both these phases, estrogens acting via estrogen receptor-α in osteoblast lineage cells are crucial for male cortical and trabecular bone, as evident from conditional genetic mouse models, epidemiological studies, rare genetic conditions, genome-wide meta-analyses, and recent interventional trials. Genetic mouse models have also demonstrated a direct role for androgens independent of aromatization on trabecular bone via the androgen receptor in osteoblasts and osteocytes, although the target cell for their key effects on periosteal bone formation remains elusive. Low serum estradiol predicts incident fractures, but the highest risk occurs in men with additionally low T and high SHBG. Still, the possible clinical utility of serum sex steroids for fracture prediction is unknown. It is likely that sex steroid actions on male bone metabolism rely also on extraskeletal mechanisms and cross talk with other signaling pathways. We propose that estrogens influence fracture risk in aging men via direct effects on bone, whereas androgens exert an additional antifracture effect mainly via extraskeletal parameters such as muscle mass and propensity to fall. Given the demographic trends of increased longevity and consequent rise of osteoporosis, an increased understanding of how sex steroids influence male bone health remains a high research priority.
Collapse
Affiliation(s)
- Dirk Vanderschueren
- Clinical and Experimental Endocrinology (D.V.) and Gerontology and Geriatrics (M.R.L., E.G.), Department of Clinical and Experimental Medicine; Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine (M.R.L., F.C.); and Centre for Metabolic Bone Diseases (D.V., M.R.L., E.G.), KU Leuven, B-3000 Leuven, Belgium; and Center for Bone and Arthritis Research (M.K.L., L.V., A.E.B., C.O.), Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Devlin MJ, Van Vliet M, Motyl K, Karim L, Brooks DJ, Louis L, Conlon C, Rosen CJ, Bouxsein ML. Early-onset type 2 diabetes impairs skeletal acquisition in the male TALLYHO/JngJ mouse. Endocrinology 2014; 155:3806-16. [PMID: 25051433 PMCID: PMC4164927 DOI: 10.1210/en.2014-1041] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Type 2 diabetes (T2D) incidence in adolescents is rising and may interfere with peak bone mass acquisition. We tested the effects of early-onset T2D on bone mass, microarchitecture, and strength in the TALLYHO/JngJ mouse, which develops T2D by 8 weeks of age. We assessed metabolism and skeletal acquisition in male TALLYHO/JngJ and SWR/J controls (n = 8-10/group) from 4 weeks to 8 and 17 weeks of age. Tallyho mice were obese; had an approximately 2-fold higher leptin and percentage body fat; and had lower bone mineral density vs SWR at all time points (P < .03 for all). Tallyho had severe deficits in distal femur trabecular bone volume fraction (-54%), trabecular number (-27%), and connectivity density (-82%) (P < .01 for all). Bone formation was higher in Tallyho mice at 8 weeks but lower by 17 weeks of age vs SWR despite similar numbers of osteoblasts. Bone marrow adiposity was 7- to 50-fold higher in Tallyho vs SWR. In vitro, primary bone marrow stromal cell differentiation into osteoblast and adipocyte lineages was similar in SWR and Tallyho, suggesting skeletal deficits were not due to intrinsic defects in Tallyho bone-forming cells. These data suggest the Tallyho mouse might be a useful model to study the skeletal effects of adolescent T2D.
Collapse
Affiliation(s)
- M J Devlin
- Department of Anthropology (M.J.D.), University of Michigan, Ann Arbor, Michigan 48104; Center for Advanced Orthopedic Studies (M.J.D., M.V.V., L.K., D.J.B., L.L., C.C., M.L.B.), Beth Israel Deaconess Medical Center, and Harvard Medical School (M.L.B.), Boston, Massachusetts 02215; and Maine Medical Center Research Institute (K.M., C.J.R.), Scarborough, Maine 04074
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Swift JM, Swift SN, Allen MR, Bloomfield SA. Beta-1 adrenergic agonist treatment mitigates negative changes in cancellous bone microarchitecture and inhibits osteocyte apoptosis during disuse. PLoS One 2014; 9:e106904. [PMID: 25211027 PMCID: PMC4161377 DOI: 10.1371/journal.pone.0106904] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 08/06/2014] [Indexed: 11/23/2022] Open
Abstract
The sympathetic nervous system (SNS) plays an important role in mediating bone remodeling. However, the exact role that beta-1 adrenergic receptors (beta1AR) have in this process has not been elucidated. We have previously demonstrated the ability of dobutamine (DOB), primarily a beta1AR agonist, to inhibit reductions in cancellous bone formation and mitigate disuse-induced loss of bone mass. The purpose of this study was to characterize the independent and combined effects of DOB and hindlimb unloading (HU) on cancellous bone microarchitecture, tissue-level bone cell activity, and osteocyte apoptosis. Male Sprague-Dawley rats, aged 6-mos, were assigned to either normal cage activity (CC) or HU (n = 18/group) for 28 days. Animals were administered either daily DOB (4 mg/kg BW/d) or an equal volume of saline (VEH) (n = 9/gp). Unloading resulted in significantly lower distal femur cancellous BV/TV (−33%), Tb.Th (−11%), and Tb.N (−25%) compared to ambulatory controls (CC-VEH). DOB treatment during HU attenuated these changes in cancellous bone microarchitecture, resulting in greater BV/TV (+29%), Tb.Th (+7%), and Tb.N (+21%) vs. HU-VEH. Distal femur cancellous vBMD (+11%) and total BMC (+8%) were significantly greater in DOB- vs. VEH-treated unloaded rats. Administration of DOB during HU resulted in significantly greater osteoid surface (+158%) and osteoblast surface (+110%) vs. HU-VEH group. Furthermore, Oc.S/BS was significantly greater in HU-DOB (+55%) vs. CC-DOB group. DOB treatment during unloading fully restored bone formation, resulting in significantly greater bone formation rate (+200%) than in HU-VEH rats. HU resulted in an increased percentage of apoptotic cancellous osteocytes (+85%), reduced osteocyte number (−16%), lower percentage of occupied osteocytic lacunae (−30%) as compared to CC-VEH, these parameters were all normalized with DOB treatment. Altogether, these data indicate that beta1AR agonist treatment during disuse mitigates negative changes in cancellous bone microarchitecture and inhibits increases in osteocyte apoptosis.
Collapse
Affiliation(s)
- Joshua M. Swift
- Departments of Health and Kinesiology, Texas A & M University, College Station, Texas, United States of America
- * E-mail:
| | - Sibyl N. Swift
- Intercollegiate Faculty of Nutrition, Texas A & M University, College Station, Texas, United States of America
| | - Matthew R. Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine (IUSM), Indianapolis, Indiana, United States of America
| | - Susan A. Bloomfield
- Departments of Health and Kinesiology, Texas A & M University, College Station, Texas, United States of America
- Intercollegiate Faculty of Nutrition, Texas A & M University, College Station, Texas, United States of America
| |
Collapse
|
35
|
Elefteriou F, Campbell P, Ma Y. Control of bone remodeling by the peripheral sympathetic nervous system. Calcif Tissue Int 2014; 94:140-51. [PMID: 23765388 PMCID: PMC3883940 DOI: 10.1007/s00223-013-9752-4] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 05/15/2013] [Indexed: 12/21/2022]
Abstract
The skeleton is no longer seen as a static, isolated, and mostly structural organ. Over the last two decades, a more complete picture of the multiple functions of the skeleton has emerged, and its interactions with a growing number of apparently unrelated organs have become evident. The skeleton not only reacts to mechanical loading and inflammatory, hormonal, and mineral challenges, but also acts of its own accord by secreting factors controlling the function of other tissues, including the kidney and possibly the pancreas and gonads. It is thus becoming widely recognized that it is by nature an endocrine organ, in addition to a structural organ and site of mineral storage and hematopoiesis. Consequently and by definition, bone homeostasis must be tightly regulated and integrated with the biology of other organs to maintain whole body homeostasis, and data uncovering the involvement of the central nervous system (CNS) in the control of bone remodeling support this concept. The sympathetic nervous system (SNS) represents one of the main links between the CNS and the skeleton, based on a number of anatomic, pharmacologic, and genetic studies focused on β-adrenergic receptor (βAR) signaling in bone cells. The goal of this report was to review the data supporting the role of the SNS and βAR signaling in the regulation of skeletal homeostasis.
Collapse
Affiliation(s)
- Florent Elefteriou
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA,
| | | | | |
Collapse
|
36
|
Motyl KJ, Bishop KA, DeMambro VE, Bornstein SA, Le P, Kawai M, Lotinun S, Horowitz MC, Baron R, Bouxsein ML, Rosen CJ. Altered thermogenesis and impaired bone remodeling in Misty mice. J Bone Miner Res 2013; 28:1885-97. [PMID: 23553822 PMCID: PMC3743939 DOI: 10.1002/jbmr.1943] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/04/2013] [Accepted: 03/20/2013] [Indexed: 11/08/2022]
Abstract
Fat mass may be modulated by the number of brown-like adipocytes in white adipose tissue (WAT) in humans and rodents. Bone remodeling is dependent on systemic energy metabolism and, with age, bone remodeling becomes uncoupled and brown adipose tissue (BAT) function declines. To test the interaction between BAT and bone, we employed Misty (m/m) mice, which were reported be deficient in BAT. We found that Misty mice have accelerated age-related trabecular bone loss and impaired brown fat function (including reduced temperature, lower expression of Pgc1a, and less sympathetic innervation compared to wild-type (+/ +)). Despite reduced BAT function, Misty mice had normal core body temperature, suggesting heat is produced from other sources. Indeed, upon acute cold exposure (4°C for 6 hours), inguinal WAT from Misty mice compensated for BAT dysfunction by increasing expression of Acadl, Pgc1a, Dio2, and other thermogenic genes. Interestingly, acute cold exposure also decreased Runx2 and increased Rankl expression in Misty bone, but only Runx2 was decreased in wild-type. Browning of WAT is under the control of the sympathetic nervous system (SNS) and, if present at room temperature, could impact bone metabolism. To test whether SNS activity could be responsible for accelerated trabecular bone loss, we treated wild-type and Misty mice with the β-blocker, propranolol. As predicted, propranolol slowed trabecular bone volume/total volume (BV/TV) loss in the distal femur of Misty mice without affecting wild-type. Finally, the Misty mutation (a truncation of DOCK7) also has a significant cell-autonomous role. We found DOCK7 expression in whole bone and osteoblasts. Primary osteoblast differentiation from Misty calvaria was impaired, demonstrating a novel role for DOCK7 in bone remodeling. Despite the multifaceted effects of the Misty mutation, we have shown that impaired brown fat function leads to altered SNS activity and bone loss, and for the first time that cold exposure negatively affects bone remodeling.
Collapse
Affiliation(s)
- Katherine J Motyl
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, ME, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Devlin MJ, Grasemann C, Cloutier AM, Louis L, Alm C, Palmert MR, Bouxsein ML. Maternal perinatal diet induces developmental programming of bone architecture. J Endocrinol 2013; 217:69-81. [PMID: 23503967 PMCID: PMC3792707 DOI: 10.1530/joe-12-0403] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Maternal high-fat (HF) diet can alter offspring metabolism via perinatal developmental programming. This study tests the hypothesis that maternal HF diet also induces perinatal programming of offspring bone mass and strength. We compared skeletal acquisition in pups from C57Bl/6J mice fed HF or normal diet from preconception through lactation. Three-week-old male and female pups from HF (HF-N) and normal mothers (N-N) were weaned onto normal diet. Outcomes at 14 and 26 weeks of age included body mass, body composition, whole-body bone mineral content (WBBMC) via peripheral dual-energy X-ray absorptiometry, femoral cortical and trabecular architecture via microcomputed tomography, and glucose tolerance. Female HF-N had normal body mass and glucose tolerance, with lower body fat (%) but higher serum leptin at 14 weeks vs. N-N (P<0.05 for both). WBBMC was 12% lower at 14 weeks and 5% lower at 26 weeks, but trabecular bone volume fraction was 20% higher at 14 weeks in female HF-N vs. N-N (P<0.05 for all). Male HF-N had normal body mass and mildly impaired glucose tolerance, with lower body fat (%) at 14 weeks and lower serum leptin at 26 weeks vs. N-N (P<0.05 for both). Serum insulin was higher at 14 weeks and lower at 26 weeks in HF-N vs. N-N (P<0.05). Trabecular BV/TV was 34% higher and cortical bone area was 6% higher at 14 weeks vs. N-N (P<0.05 for both). These data suggest that maternal HF diet has complex effects on offspring bone, supporting the hypothesis that maternal diet alters postnatal skeletal homeostasis.
Collapse
Affiliation(s)
- M J Devlin
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Ji-Ye H, Xin-Feng Z, Lei-Sheng J. Autonomic control of bone formation. AUTONOMIC NERVOUS SYSTEM 2013; 117:161-71. [DOI: 10.1016/b978-0-444-53491-0.00014-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
39
|
|
40
|
Farr JN, Charkoudian N, Barnes JN, Monroe DG, McCready LK, Atkinson EJ, Amin S, Melton LJ, Joyner MJ, Khosla S. Relationship of sympathetic activity to bone microstructure, turnover, and plasma osteopontin levels in women. J Clin Endocrinol Metab 2012; 97:4219-27. [PMID: 22948767 PMCID: PMC3485606 DOI: 10.1210/jc.2012-2381] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
CONTEXT Studies in rodents have demonstrated that sympathetic activity reduces bone formation and bone mass; these effects are mediated by the noncollagenous matrix protein, osteopontin. OBJECTIVE The objective of the study was to relate sympathetic activity (measured using microneurography at the peroneal nerve) to bone microstructure (assessed by high resolution peripheral quantitative computed tomography), bone turnover, and plasma osteopontin levels. DESIGN, SETTING, AND PATIENTS Twenty-three women aged 20-72 yr (10 premenopausal and 13 postmenopausal) were studied in the Clinical Research Unit. RESULTS Sympathetic activity (bursts per 100 heart beats) was 2.4-fold higher in postmenopausal as compared with premenopausal women (P < 0.001). In the two groups combined and after age adjustment, sympathetic activity was inversely correlated with trabecular bone volume fraction (r = -0.55, P < 0.01) and thickness (r = -0.59, P < 0.01) and positively correlated with trabecular separation (r = 0.45, P < 0.05). Sympathetic activity was negatively correlated with serum amino-terminal propeptide of type I collagen in postmenopausal women (r = -0.65, P = 0.015), with a similar trend in premenopausal women (r = -0.58, P = 0.082). Sympathetic activity was also negatively correlated with plasma osteopontin levels (r = -0.43, P = 0.045), driven mainly by the correlation in postmenopausal women (r = -0.76, P = 0.002). CONCLUSION These findings represent the first demonstration in humans of a relationship between sympathetic activity and bone microstructure and circulating levels of amino-terminal propeptide of type I collagen and osteopontin. Given the critical role of osteopontin in mediating the effects of β-adrenergic signaling on bone, the inverse association between sympathetic activity and plasma osteopontin levels may reflect a negative feedback loop to limit the deleterious effects of sympathetic activity on bone metabolism. Based on the higher sympathetic activity observed in postmenopausal women, additional human studies are needed to define the role of increased sympathetic activity in mediating postmenopausal bone loss.
Collapse
Affiliation(s)
- Joshua N Farr
- College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Grskovic I, Kutsch A, Frie C, Groma G, Stermann J, Schlötzer-Schrehardt U, Niehoff A, Moss SE, Rosenbaum S, Pöschl E, Chmielewski M, Rappl G, Abken H, Bateman JF, Cheah KS, Paulsson M, Brachvogel B. Depletion of annexin A5, annexin A6, and collagen X causes no gross changes in matrix vesicle-mediated mineralization, but lack of collagen X affects hematopoiesis and the Th1/Th2 response. J Bone Miner Res 2012; 27:2399-412. [PMID: 22692895 DOI: 10.1002/jbmr.1682] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Numerous biochemical studies have pointed to an essential role of annexin A5 (AnxA5), annexin A6 (AnxA6), and collagen X in matrix vesicle-mediated biomineralization during endochondral ossification and in osteoarthritis. By binding to the extracellular matrix protein collagen X and matrix vesicles, annexins were proposed to anchor matrix vesicles in the extracellular space of hypertrophic chondrocytes to initiate the calcification of cartilage. However, mineralization appears to be normal in mice lacking AnxA5 and AnxA6, whereas collagen X-deficient mice show only subtle alterations in the growth plate organization. We hypothesized that the simultaneous lack of AnxA5, AnxA6, and collagen X in vivo induces more pronounced changes in the growth plate development and the initiation of mineralization. In this study, we generated and analyzed mice deficient for AnxA5, AnxA6, and collagen X. Surprisingly, mice were viable, fertile, and showed no obvious abnormalities. Assessment of growth plate development indicated that the hypertrophic zone was expanded in Col10a1(-/-) and AnxA5(-/-) AnxA6(-/-) Col10a1(-/-) newborns, whereas endochondral ossification and mineralization were not affected in 13-day- and 1-month-old mutants. In peripheral quantitative computed tomography, no changes in the degree of biomineralization were found in femora of 1-month- and 1-year-old mutants even though the diaphyseal circumference was reduced in Col10a1(-/-) and AnxA5(-/-) AnxA6(-/-) Col10a1(-/-) mice. The percentage of naive immature IgM(+) /IgM(+) B cells and peripheral T-helper cells were increased in Col10a1(-/-) and AnxA5(-/-) AnxA6(-/-) Col10a1(-/-) mutants, and activated splenic T cells isolated from Col10a1(-/-) mice secreted elevated levels of IL-4 and GM-CSF. Hence, collagen X is needed for hematopoiesis during endochondral ossification and for the immune response, but the interaction of annexin A5, annexin A6, and collagen X is not essential for physiological calcification of growth plate cartilage. Therefore, annexins and collagen X may rather fulfill functions in growth plate cartilage not directly linked to the mineralization process.
Collapse
Affiliation(s)
- Ivan Grskovic
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Camerino C, Zayzafoon M, Rymaszewski M, Heiny J, Rios M, Hauschka PV. Central depletion of brain-derived neurotrophic factor in mice results in high bone mass and metabolic phenotype. Endocrinology 2012; 153:5394-405. [PMID: 23011922 PMCID: PMC3685798 DOI: 10.1210/en.2012-1378] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) plays important roles in neuronal differentiation/survival, the regulation of food intake, and the pathobiology of obesity and type 2 diabetes mellitus. BDNF and its receptor are expressed in osteoblasts and chondrocyte. BDNF in vitro has a positive effect on bone; whether central BDNF affects bone mass in vivo is not known. We therefore examined bone mass and energy use in brain-targeted BDNF conditional knockout mice (Bdnf(2lox/2lox)/93). The deletion of BDNF in the brain led to a metabolic phenotype characterized by hyperphagia, obesity, and increased abdominal white adipose tissue. Central BDNF deletion produces a marked skeletal phenotype characterized by increased femur length, elevated whole bone mineral density, and bone mineral content. The skeletal changes are developmentally regulated and appear concurrently with the metabolic phenotype, suggesting that the metabolic and skeletal actions of BDNF are linked. The increased bone development is evident in both the cortical and trabecular regions. Compared with control, Bdnf(2lox/2lox)/93 mice show greater trabecular bone volume (+50% for distal femur, P < 0.001; +35% for vertebral body, P < 0.001) and midfemoral cortical thickness (+11 to 17%, P < 0.05), measured at 3 and 6 months of age. The skeletal and metabolic phenotypes were gender dependent, with female being more affected than male mice. However, uncoupling protein-1 expression in brown fat, a marker of sympathetic tone, was not different between genotypes. We show that deletion of central BDNF expression in mice results in increased bone mass and white adipose tissue, with no significant changes in sympathetic signaling or peripheral serotonin, associated with hyperphagia, obesity, and leptin resistance.
Collapse
Affiliation(s)
- C Camerino
- Harvard School of Dental Medicine, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Long-term propranolol use in severely burned pediatric patients: a randomized controlled study. Ann Surg 2012; 256:402-11. [PMID: 22895351 DOI: 10.1097/sla.0b013e318265427e] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To determine the safety and efficacy of propranolol given for 1 year on cardiac function, resting energy expenditure, and body composition in a prospective, randomized, single-center, controlled study in pediatric patients with large burns. BACKGROUND Severe burns trigger a hypermetabolic response that persists for up to 2 years postburn. Propranolol given for 1 month postburn blunts this response. Whether propranolol administration for 1 year after injury provides a continued benefit is currently unclear. METHODS One-hundred seventy-nine pediatric patients with more than 30% total body surface area burns were randomized to control (n = 89) or 4 mg/kg/d propranolol (n = 90) for 12 months postburn. Changes in resting energy expenditure, cardiac function, and body composition were measured acutely at 3, 6, 9, and 12 months postburn. Statistical analyses included techniques that adjusted for non-normality, repeated-measures, and regression analyses. P < 0.05 was considered significant. RESULTS Long-term propranolol treatment significantly reduced the percentage of the predicted heart rate and percentage of the predicted resting energy expenditure, decreased accumulation of central mass and central fat, prevented bone loss, and improved lean body mass accretion. There were very few adverse effects from the dose of propranolol used. CONCLUSIONS Propranolol treatment for 12 months after thermal injury, ameliorates the hyperdynamic, hypermetabolic, hypercatabolic, and osteopenic responses in pediatric patients. This study is registered at clinicaltrials.gov: NCT00675714.
Collapse
|
44
|
Pierroz DD, Bonnet N, Bianchi EN, Bouxsein ML, Baldock PA, Rizzoli R, Ferrari SL. Deletion of β-adrenergic receptor 1, 2, or both leads to different bone phenotypes and response to mechanical stimulation. J Bone Miner Res 2012; 27:1252-62. [PMID: 22407956 DOI: 10.1002/jbmr.1594] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
As they age, mice deficient for the β2-adrenergic receptor (Adrb2(-/-) ) maintain greater trabecular bone microarchitecture, as a result of lower bone resorption and increased bone formation. The role of β1-adrenergic receptor signaling and its interaction with β2-adrenergic receptor on bone mass regulation, however, remains poorly understood. We first investigated the skeletal response to mechanical stimulation in mice deficient for β1-adrenergic receptors and/or β2-adrenergic receptors. Upon axial compression loading of the tibia, bone density, cancellous and cortical microarchitecture, as well as histomorphometric bone forming indices, were increased in both Adrb2(-/-) and wild-type (WT) mice, but not in Adrb1(-/-) nor in Adrb1b2(-/-) mice. Moreover, in the unstimulated femur and vertebra, bone mass and microarchitecture were increased in Adrb2(-/-) mice, whereas in Adrb1(-/-) and Adrb1b2(-/-) double knockout mice, femur bone mineral density (BMD), cancellous bone volume/total volume (BV/TV), cortical size, and cortical thickness were lower compared to WT. Bone histomorphometry and biochemical markers showed markedly decreased bone formation in Adrb1b2(-/-) mice during growth, which paralleled a significant decline in circulating insulin-like growth factor 1 (IGF-1) and IGF-binding protein 3 (IGF-BP3). Finally, administration of the β-adrenergic agonist isoproterenol increased bone resorption and receptor activator of NF-κB ligand (RANKL) and decreased bone mass and microarchitecture in WT but not in Adrb1b2(-/-) mice. Altogether, these results demonstrate that β1- and β2-adrenergic signaling exert opposite effects on bone, with β1 exerting a predominant anabolic stimulus in response to mechanical stimulation and during growth, whereas β2-adrenergic receptor signaling mainly regulates bone resorption during aging.
Collapse
Affiliation(s)
- Dominique D Pierroz
- Service of Bone Diseases, Department of Rehabilitation and Geriatrics, Geneva University Hospital and Faculty of Medicine, Geneva, Switzerland
| | | | | | | | | | | | | |
Collapse
|
45
|
Bataille C, Mauprivez C, Haÿ E, Baroukh B, Brun A, Chaussain C, Marie PJ, Saffar JL, Cherruau M. Different sympathetic pathways control the metabolism of distinct bone envelopes. Bone 2012; 50:1162-72. [PMID: 22326888 DOI: 10.1016/j.bone.2012.01.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 01/16/2012] [Accepted: 01/25/2012] [Indexed: 10/14/2022]
Abstract
Bone remodeling, the mechanism that modulates bone mass adaptation, is controlled by the sympathetic nervous system through the catecholaminergic pathway. However, resorption in the mandible periosteum envelope is associated with cholinergic Vasoactive Intestinal Peptide (VIP)-positive nerve fibers sensitive to sympathetic neurotoxics, suggesting that different sympathetic pathways may control distinct bone envelopes. In this study, we assessed the role of distinct sympathetic pathways on rat femur and mandible envelopes. To this goal, adult male Wistar rats were chemically sympathectomized or treated with agonists/antagonists of the catecholaminergic and cholinergic pathways; femora and mandibles were sampled. Histomorphometric analysis showed that sympathectomy decreased the number of preosteoclasts and RANKL-expressing osteoblasts in mandible periosteum but had no effect on femur trabecular bone. In contrast, pharmacological stimulation or repression of the catecholaminergic cell receptors impacted the femur trabecular bone and mandible endosteal retromolar zone. VIP treatment of sympathectomized rats rescued the disturbances of the mandible periosteum and alveolar wall whereas the cholinergic pathway had no effect on the catecholaminergic-dependent envelopes. We also found that VIP receptor-1 was weakly expressed in periosteal osteoblasts in the mandible and was increased by VIP treatment, whereas osteoblasts of the retromolar envelope that was innervated only by tyrosine hydroxylase-immunoreactive fibers, constitutively expressed beta-2 adrenergic receptors. These data highlight the complexity of the sympathetic control of bone metabolism. Both the embryological origin of the bone (endochondral for the femur, membranous for the mandibular periosteum and the socket wall) and environmental factors specific to the innervated envelope may influence the phenotype of the sympathetic innervation. We suggest that an origin-dependent imprint of bone cells through osteoblast-nerve interactions determines the type of autonomous system innervating a particular bone envelope.
Collapse
Affiliation(s)
- Caroline Bataille
- EA2496 Laboratoire Pathologies et Biothérapies de l'Organe Dentaire, Faculté de Chirurgie Dentaire, Université Paris Descartes, Sorbonne Paris Cité, 1 rue Maurice Arnoux 92120 Montrouge, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Effects of propranolol on the development of glucocorticoid-induced osteoporosis in male rats. Pharmacol Rep 2012; 63:1040-9. [PMID: 22001992 DOI: 10.1016/s1734-1140(11)70620-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 04/14/2011] [Indexed: 02/07/2023]
Abstract
Glucocorticoid-induced osteoporosis is the most frequently occurring type of secondary osteoporosis. Antagonists of β-adrenergic receptors are now considered to be potential drugs under investigation for osteoporosis. The aim of the present study was to investigate the effects of propranolol, a nonselective β-receptor antagonist, on the skeletal system of mature male rats and on the development of bone changes induced by glucocorticoid (prednisolone) administration. The experiments were performed on 24-week-old male Wistar rats. The effects of prednisolone 21-hemisuccinate sodium salt (7 mg/kg, sc daily) or/and propranolol hydrochloride (10 mg/kg, ip daily) administered for 4 weeks on the skeletal system were studied. Bone and bone mineral mass in the tibia, femur and L-4 vertebra, length and diameter of the long bones, mechanical properties of tibial metaphysis, femoral diaphysis and femoral neck, bone histomorphometric parameters and turnover markers in serum were determined. Prednisolone-induced unfavorable skeletal changes led to disorders in bone mechanical properties. Propranolol not only did not improve bone parameters, but even caused deleterious effects on the skeletal system. Concurrent administration of propranolol with prednisolone did not counteract the changes induced by prednisolone. The results of this study may help to understand the equivocal results of human studies on the effects of β-blockers on the skeletal system. It is possible that the drugs exert biphasic effects on the skeletal system, both favorable and deleterious, depending on the dose or individual susceptibility.
Collapse
|
47
|
Lecka-Czernik B. Marrow fat metabolism is linked to the systemic energy metabolism. Bone 2012; 50:534-9. [PMID: 21757043 PMCID: PMC3197966 DOI: 10.1016/j.bone.2011.06.032] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 06/24/2011] [Accepted: 06/25/2011] [Indexed: 12/25/2022]
Abstract
Recent advances in understanding the role of bone in the systemic regulation of energy metabolism indicate that bone marrow cells, adipocytes and osteoblasts, are involved in this process. Marrow adipocytes store significant quantities of fat and produce adipokines, leptin and adiponectin, which are known for their role in the regulation of energy metabolism, whereas osteoblasts produce osteocalcin, a bone-specific hormone that has a potential to regulate insulin production in the pancreas and adiponectin production in fat tissue. Both osteoblasts and marrow adipocytes express insulin receptor and respond to insulin-sensitizing anti-diabetic TZDs in a manner, which tightly links bone with the energy metabolism system. Metabolic profile of marrow fat resembles that of both, white and brown fat, which is reflected by its plasticity in acquiring different functions including maintenance of bone micro-environment. Marrow fat responds to physiologic and pathologic changes in energy metabolism status by changing volume and metabolic activity. This review summarizes available information on the metabolic function of marrow fat and provides hypothesis that this fat depot may acquire multiple roles depending on the local and perhaps systemic demands. These functions may include a role in bone energy maintenance and endocrine activities to serve osteogenesis during bone remodeling and bone healing.
Collapse
Affiliation(s)
- Beata Lecka-Czernik
- Department of Orthopaedic Surgery, University of Toledo Health Sciences Campus, Toledo, OH 43614, USA.
| |
Collapse
|
48
|
Cheung CL, Sham PC, Xiao SM, Bow CH, Kung AWC. Meta-analysis of gene-based genome-wide association studies of bone mineral density in Chinese and European subjects. Osteoporos Int 2012; 23:131-42. [PMID: 21927923 PMCID: PMC3249198 DOI: 10.1007/s00198-011-1779-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 07/05/2011] [Indexed: 12/19/2022]
Abstract
UNLABELLED Gene-based association approach could be regarded as a complementary analysis to the single SNP association analysis. We meta-analyzed the findings from the gene-based association approach using the genome-wide association studies (GWAS) data from Chinese and European subjects, confirmed several well established bone mineral density (BMD) genes, and suggested several novel BMD genes. INTRODUCTION The introduction of GWAS has greatly increased the number of genes that are known to be associated with common diseases. Nonetheless, such a single SNP GWAS has a lower power to detect genes with multiple causal variants. We aimed to assess the association of each gene with BMD variation at the spine and hip using gene-based GWAS approach. METHODS We studied 778 Hong Kong Southern Chinese (HKSC) women and 5,858 Northern Europeans (dCG); age, sex, and weight were adjusted in the model. The main outcome measure was BMD at the spine and hip. RESULTS Nine genes showed suggestive p value in HKSC, while 4 and 17 genes showed significant and suggestive p values respectively in dCG. Meta-analysis using weighted Z-transformed test confirmed several known BMD genes and suggested some novel ones at 1q21.3, 9q22, 9q33.2, 20p13, and 20q12. Top BMD genes were significantly associated with connective tissue, skeletal, and muscular system development and function (p < 0.05). Gene network inference revealed that a large number of these genes were significantly connected with each other to form a functional gene network, and several signaling pathways were strongly connected with these gene networks. CONCLUSION Our gene-based GWAS confirmed several BMD genes and suggested several novel BMD genes. Genetic contribution to BMD variation may operate through multiple genes identified in this study in functional gene networks. This finding may be useful in identifying and prioritizing candidate genes/loci for further study.
Collapse
Affiliation(s)
- C-L Cheung
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, China.
| | | | | | | | | |
Collapse
|
49
|
He JY, Jiang LS, Dai LY. The roles of the sympathetic nervous system in osteoporotic diseases: A review of experimental and clinical studies. Ageing Res Rev 2011; 10:253-63. [PMID: 21262391 DOI: 10.1016/j.arr.2011.01.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2010] [Revised: 01/10/2011] [Accepted: 01/11/2011] [Indexed: 02/04/2023]
Abstract
With the rapid aging of the world population, the issue of skeletal health is becoming more prominent and urgent. The bone remodeling mechanism has sparked great interest among bone research societies. At the same time, increasing clinical and experimental evidence has driven attention towards the pivotal role of the sympathetic nervous system (SNS) in bone remodeling. Bone remodeling is thought to be partially controlled by the hypothalamus, a process which is mediated by the adrenergic nerves and neurotransmitters. Currently, new knowledge about the role of the SNS in the development and pathophysiology of osteoporosis is being generated. The aim of this review is to summarize the evidence that proves the involvement of the SNS in bone metabolism and to outline some common osteoporotic diseases that occur under different circumstances. The adrenergic signaling pathway and its neurotransmitters are involved to various degrees of importance in the development of osteoporosis in postmenopause, as well as in spinal cord injury, depression, unloading and the complex regional pain syndrome. In addition, clinical and pharmacological studies have helped to increase the comprehension of the adrenergic signaling pathway. We try to individually examine the contributions of the SNS in osteoporotic diseases from a different perspective. It is our hope that a further understanding of the adrenergic signaling by the SNS will pave the way for conceptualizing optimal treatment regimens for osteoporosis in the near future.
Collapse
Affiliation(s)
- Ji-Ye He
- Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, China
| | | | | |
Collapse
|
50
|
Motyl KJ, Rosen CJ. Temperatures rising: brown fat and bone. DISCOVERY MEDICINE 2011; 11:179-185. [PMID: 21447277 PMCID: PMC3629549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Caloric restriction is associated with a reduction in body weight and temperature, as well as a reduction in trabecular bone volume and paradoxically an increase in adipocytes within the bone marrow. The nature of these adipocytes is uncertain, although there is emerging evidence of a direct relationship between bone remodeling and brown adipocytes. For example, in heterotrophic ossification, brown adipocytes set up a hypoxic gradient that leads to vascular invasion, chondrocyte differentiation, and subsequent bone formation. Additionally, deletion of retinoblastoma protein in an osteosarcoma model leads to increased hibernoma (brown fat tumor). Brown adipose tissue (BAT) becomes senescent with age at a time when thermoregulation is altered, bone loss becomes apparent, and sympathetic activity increases. Interestingly, heart rate is an unexpected but good predictor of fracture risk in elderly individuals, pointing to a key role for the sympathetic nervous system in senile osteoporosis. Hence the possibility exists that BAT could play an indirect role in age-related bone loss. However, evidence of an indirect effect from thermogenic dysfunction on bone loss is currently limited. Here, we present current evidence for a relationship between brown adipose tissue and bone as well as provide novel insights into the effects of thermoregulation on bone mineral density.
Collapse
Affiliation(s)
- Katherine J. Motyl
- Maine Medical Center Research Institute 81 Research Drive Scarborough, ME 04074
| | - Clifford J. Rosen
- Maine Medical Center Research Institute 81 Research Drive Scarborough, ME 04074
| |
Collapse
|