1
|
Marino S, Bellido T. PTH receptor signalling, osteocytes and bone disease induced by diabetes mellitus. Nat Rev Endocrinol 2024; 20:661-672. [PMID: 39020007 DOI: 10.1038/s41574-024-01014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 07/19/2024]
Abstract
Basic, translational and clinical research over the past few decades has provided new understanding on the mechanisms by which activation of the receptor of parathyroid hormone (parathyroid hormone 1 receptor (PTH1R)) regulates bone physiology and pathophysiology. A fundamental change in the field emerged upon the recognition that osteocytes, which are permanent residents of bone and the most abundant cells in bone, are targets of the actions of natural and synthetic ligands of PTH1R (parathyroid hormone and abaloparatide, respectively), and that these cells drive essential actions related to bone remodelling. Among the numerous genes regulated by PTH1R in osteocytes, SOST (which encodes sclerostin, the WNT signalling antagonist and inhibitor of bone formation) has a critical role in bone homeostasis and changes in its expression are associated with several bone pathologies. The bone fragility syndrome induced by diabetes mellitus is accompanied by increased osteocyte apoptosis and changes in the expression of osteocytic genes, including SOST. This Review will discuss advances in our knowledge of the role of osteocytes in PTH1R signalling and the new opportunities to restore bone health in diabetes mellitus by targeting the osteocytic PTH1R-sclerostin axis.
Collapse
Affiliation(s)
- Silvia Marino
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Central Arkansas Veterans Healthcare System, John L. McClellan Little Rock, Little Rock, AR, USA
| | - Teresita Bellido
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
- Central Arkansas Veterans Healthcare System, John L. McClellan Little Rock, Little Rock, AR, USA.
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
2
|
Abe M, Hasegawa T, Hongo H, Yamamoto T, Shi Y, Cui J, Liu X, Yao Q, Ishizu H, Maruoka H, Yoshino H, Haraguchi-Kitakamae M, Shimizu T, Amizuka N. Immunohistochemical and Morphometric Assessment on the Biological Function and Vascular Endothelial Cells in the Initial Process of Cortical Porosity in Mice With PTH Administration. J Histochem Cytochem 2024; 72:309-327. [PMID: 38725403 PMCID: PMC11107436 DOI: 10.1369/00221554241247883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 03/29/2024] [Indexed: 05/18/2024] Open
Abstract
To clarify the cellular mechanism of cortical porosity induced by intermittent parathyroid hormone (PTH) administration, we examined the femoral cortical bone of mice that received 40 µg/kg/day (four times a day) human PTH (hPTH) (1-34). The PTH-driven cortical porosity initiated from the metaphyseal region and chronologically expanded toward the diaphysis. Alkaline phosphatase (ALP)-positive osteoblasts in the control mice covered the cortical surface, and endomucin-positive blood vessels were distant from these osteoblasts. In PTH-administered mice, endomucin-reactive blood vessels with TRAP-positive penetrated the ALP-positive osteoblast layer, invading the cortical bone. Statistically, the distance between endomucin-positive blood vessels and the cortical bone surface abated after PTH administration. Transmission electron microscopic observation demonstrated that vascular endothelial cells often pass through the flattened osteoblast layer and accompanied osteoclasts in the deep region of the cortical bone. The cell layers covering mature osteoblasts thickened with PTH administration and exhibited ALP, α-smooth muscle actin (αSMA), vascular cell adhesion molecule-1 (VCAM1), and receptor activator of NF-κB ligand (RANKL). Within these cell layers, osteoclasts were found near endomucin-reactive blood vessels. In PTH-administered femora, osteocytes secreted Dkk1, a Wnt inhibitor that affects angiogenesis, and blood vessels exhibited plasmalemma vesicle-associated protein, an angiogenic molecule. In summary, endomucin-positive blood vessels, when accompanied by osteoclasts in the ALP/αSMA/VCAM1/RANKL-reactive osteoblastic cell layers, invade the cortical bone, potentially due to the action of osteocyte-derived molecules such as DKK1.
Collapse
Affiliation(s)
- Miki Abe
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| | - Tomoka Hasegawa
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| | - Hiromi Hongo
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| | - Tomomaya Yamamoto
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
- Hokkaido University, Sapporo, Japan, and Department of Dentistry, Japan Ground Self-Defense Force Camp Shinmachi, Takasaki, Japan
| | - Yan Shi
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| | - Jiaxin Cui
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| | - Xuanyu Liu
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| | - Qi Yao
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| | - Hotaka Ishizu
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine and Orthopedics, Graduate School of Medicine, Faculty of Medicine
| | - Haruhi Maruoka
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| | - Hirona Yoshino
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| | - Mai Haraguchi-Kitakamae
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| | | | - Norio Amizuka
- Ultrastructure of Hard Tissue, Graduate School of Dental Medicine, Faculty of Dental Medicine
| |
Collapse
|
3
|
Hannachi M, Ouerghi N, Abassi W, Ouergui I, kammoun I, Bouassida A. Effects of high- vs. moderate-intensity intermittent training on parathyroid hormone concentration in overweight/obese females. Sci Sports 2023. [DOI: 10.1016/j.scispo.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
4
|
Krishnan RH, Sadu L, Akshaya RL, Gomathi K, Saranya I, Das UR, Satishkumar S, Selvamurugan N. Circ_CUX1/miR-130b-5p/p300 axis for parathyroid hormone-stimulation of Runx2 activity in rat osteoblasts: A combined bioinformatic and experimental approach. Int J Biol Macromol 2023; 225:1152-1163. [PMID: 36427609 DOI: 10.1016/j.ijbiomac.2022.11.176] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/31/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Parathyroid hormone (PTH) regulates the expression of bone remodeling genes by enhancing the activity of Runx2 in osteoblasts. p300, a histone acetyltransferase, acetylated Runx2 to activate the expression of its target genes. PTH stimulated the expression of p300 in rat osteoblastic cells. Increasing studies suggested the potential of non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and circular RNAs (circRNAs), in regulating gene expression under both physiological and pathological conditions. In this study, we hypothesized that PTH regulates Runx2 activity via ncRNAs-mediated p300 expression in rat osteoblastic cells. Bioinformatics and experimental approaches identified PTH-upregulation of miR-130b-5p and circ_CUX1 that putatively target p300 and miR-130b-5p, respectively. An antisense-mediated knockdown of circ_CUX1 was performed to determine the sponging activity of circ_CUX1. Knockdown of circ_CUX1 promoted miR-130b-5p activity and reduced p300 expression, resulting in decreased Runx2 acetylation in rat osteoblastic cells. Further, bioinformatics analysis identified the possible signaling pathways that regulate Runx2 activity and osteoblast differentiation via circ_CUX1/miR-130b-5p/p300 axis. The predicted circ_CUX1/miR-130b-5p/p300 axis might pave the way for better diagnostic and therapeutic approaches for bone-related diseases.
Collapse
Affiliation(s)
- R Hari Krishnan
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Lakshana Sadu
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - R L Akshaya
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - K Gomathi
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - I Saranya
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Udipt Ranjan Das
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Sneha Satishkumar
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
5
|
Vasiliadis ES, Evangelopoulos DS, Kaspiris A, Benetos IS, Vlachos C, Pneumaticos SG. The Role of Sclerostin in Bone Diseases. J Clin Med 2022; 11:806. [PMID: 35160258 PMCID: PMC8836457 DOI: 10.3390/jcm11030806] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/26/2022] Open
Abstract
Sclerostin has been identified as an important regulator of bone homeostasis through inhibition of the canonical Wnt-signaling pathway, and it is involved in the pathogenesis of many different skeletal diseases. Many studies have been published in the last few years regarding sclerostin's origin, regulation, and mechanism of action. The ongoing research emphasizes the potential therapeutic implications of sclerostin in many pathological conditions with or without skeletal involvement. Antisclerostin antibodies have recently been approved for the treatment of osteoporosis, and several animal studies and clinical trials are currently under way to evaluate the effectiveness of antisclerostin antibodies in the treatment of other than osteoporosis skeletal disorders and cancer with promising results. Understanding the exact role of sclerostin may lead to new therapeutic approaches for the treatment of skeletal disorders.
Collapse
Affiliation(s)
- Elias S. Vasiliadis
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Hospital, 16541 Athens, Greece; (D.-S.E.); (I.S.B.); (C.V.); (S.G.P.)
| | - Dimitrios-Stergios Evangelopoulos
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Hospital, 16541 Athens, Greece; (D.-S.E.); (I.S.B.); (C.V.); (S.G.P.)
| | - Angelos Kaspiris
- Laboratory of Molecular Pharmacology, Division for Orthopaedic Research, School of Health Sciences, University of Patras, 26504 Rion, Greece;
| | - Ioannis S. Benetos
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Hospital, 16541 Athens, Greece; (D.-S.E.); (I.S.B.); (C.V.); (S.G.P.)
| | - Christos Vlachos
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Hospital, 16541 Athens, Greece; (D.-S.E.); (I.S.B.); (C.V.); (S.G.P.)
| | - Spyros G. Pneumaticos
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Hospital, 16541 Athens, Greece; (D.-S.E.); (I.S.B.); (C.V.); (S.G.P.)
| |
Collapse
|
6
|
Xu M, Li Y, Feng X, Zheng W, Zhao Z, Li Y. Parathyroid hormone promotes maxillary expansion and reduces relapse in the repeated activation maxillary expansion rat model by regulating Wnt/β-catenin pathway. Prog Orthod 2022; 23:1. [PMID: 34978631 PMCID: PMC8724514 DOI: 10.1186/s40510-021-00394-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 11/26/2021] [Indexed: 02/08/2023] Open
Abstract
Background Constricted maxillary bone is a common skeletal deformity, which may lead to crowding and posterior crossbite. Mid-palatal suture expansion is often used to increase the maxillary width, but its skeletal effects are limited and tend to relapse, even with prolonged retention. We hypothesized that parathyroid hormone (PTH) may reduce the relapse of maxillary expansion. Methods We established a novel rat maxillary expansion model using palatal tubes with an insertable “W”-shaped spring which can be repeatedly activated. A total of 32 male healthy Wistar rats were randomly divided into six groups: the control group, the PTH group, the expansion group, the expansion + PTH group, the expansion + relapse group and the expansion + PTH + relapse group. All animals in the first 4 groups were killed after 10 days and the 2 relapse groups were killed after 15 days. The maxillary arch widths and histological staining were used to assess the expansion and relapse effects. The immunohistochemical staining, micro-CT, RT-qPCR and Western blot were used to evaluate the bone remodeling during expansion. Results The suture width was increased by the expansion device, and the repeated activation maxillary expansion rat model showed better expansion effects than the conventional model. PTH significantly promoted the expansion width and reduced the relapse ratio. Meanwhile, in the expansion + PTH group, histological and immunohistochemical staining showed that osteoblasts, osteoclasts, new cartilage and osteoid were significantly increased, micro-CT showed increased bone mass, and PCR and Western blot results confirmed up-regulation of RANKL, β-catenin, type II collagen and OCN. Conclusion The novel repeated activation maxillary expansion rat model has better effects than the conventional model. PTH enhances the maxillary expansion and reduces its relapse by regulating Wnt/β-catenin and RANKL pathways. PTH administration may serve as an adjunctive therapy in addition to mechanical expansion for treatment of maxillary constriction.
Collapse
Affiliation(s)
- Mengting Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, South Renmin Road, Chengdu, 610041, People's Republic of China
| | - Yuan Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, South Renmin Road, Chengdu, 610041, People's Republic of China
| | - Xiaoxia Feng
- The Affiliated Stomatology Hospital, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Wei Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, South Renmin Road, Chengdu, 610041, People's Republic of China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, South Renmin Road, Chengdu, 610041, People's Republic of China.
| | - Yu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, #14, 3rd Section, South Renmin Road, Chengdu, 610041, People's Republic of China
| |
Collapse
|
7
|
Abstract
Osteocytes, former osteoblasts encapsulated by mineralized bone matrix, are far from being passive and metabolically inactive bone cells. Instead, osteocytes are multifunctional and dynamic cells capable of integrating hormonal and mechanical signals and transmitting them to effector cells in bone and in distant tissues. Osteocytes are a major source of molecules that regulate bone homeostasis by integrating both mechanical cues and hormonal signals that coordinate the differentiation and function of osteoclasts and osteoblasts. Osteocyte function is altered in both rare and common bone diseases, suggesting that osteocyte dysfunction is directly involved in the pathophysiology of several disorders affecting the skeleton. Advances in osteocyte biology initiated the development of novel therapeutics interfering with osteocyte-secreted molecules. Moreover, osteocytes are targets and key distributors of biological signals mediating the beneficial effects of several bone therapeutics used in the clinic. Here we review the most recent discoveries in osteocyte biology demonstrating that osteocytes regulate bone homeostasis and bone marrow fat via paracrine signaling, influence body composition and energy metabolism via endocrine signaling, and contribute to the damaging effects of diabetes mellitus and hematologic and metastatic cancers in the skeleton.
Collapse
Affiliation(s)
- Jesus Delgado-Calle
- 1Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas,2Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Teresita Bellido
- 1Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas,2Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas,3Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| |
Collapse
|
8
|
Patil KC, Soekmadji C. Extracellular Vesicle-Mediated Bone Remodeling and Bone Metastasis: Implications in Prostate Cancer. Subcell Biochem 2021; 97:297-361. [PMID: 33779922 DOI: 10.1007/978-3-030-67171-6_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone metastasis is the tendency of certain primary tumors to spawn and dictate secondary neoplasia in the bone. The process of bone metastasis is regulated by the dynamic crosstalk between metastatic cancer cells, cellular components of the bone marrow microenvironment (osteoblasts, osteoclasts, and osteocytes), and the bone matrix. The feed-forward loop mechanisms governs the co-option of homeostatic bone remodeling by cancer cells in bone. Recent developments have highlighted the discovery of extracellular vesicles (EVs) and their diverse roles in distant outgrowths. Several studies have implicated EV-mediated interactions between cancer cells and the bone microenvironment in synergistically promoting pathological skeletal metabolism in the metastatic site. Nevertheless, the potential role that EVs serve in arbitrating intricate sequences of coordinated events within the bone microenvironment remains an emerging field. In this chapter, we review the role of cellular participants and molecular mechanisms in regulating normal bone physiology and explore the progress of current research into bone-derived EVs in directly triggering and coordinating the processes of physiological bone remodeling. In view of the emerging role of EVs in interorgan crosstalk, this review also highlights the multiple systemic pathophysiological processes orchestrated by the EVs to direct organotropism in bone in prostate cancer. Given the deleterious consequences of bone metastasis and its clinical importance, in-depth knowledge of the multifarious role of EVs in distant organ metastasis is expected to open new possibilities for prognostic evaluation and therapeutic intervention for advanced bone metastatic prostate cancer.
Collapse
Affiliation(s)
- Kalyani C Patil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia. .,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
9
|
Imerb N, Thonusin C, Chattipakorn N, Chattipakorn SC. Aging, obese-insulin resistance, and bone remodeling. Mech Ageing Dev 2020; 191:111335. [DOI: 10.1016/j.mad.2020.111335] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 08/14/2020] [Indexed: 02/08/2023]
|
10
|
Arlt H, Mullarkey T, Hu D, Baron R, Ominsky MS, Mitlak B, Lanske B, Besschetnova T. Effects of abaloparatide and teriparatide on bone resorption and bone formation in female mice. Bone Rep 2020; 13:100291. [PMID: 32637467 PMCID: PMC7330158 DOI: 10.1016/j.bonr.2020.100291] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 01/09/2023] Open
Abstract
Intermittent administration of PTH type 1 receptor (PTH1R) agonists increases bone remodeling, with greater stimulation of bone formation relative to bone resorption causing net gains in bone mass. This pharmacodynamic feature underlies the bone-building effects of teriparatide and abaloparatide, the only PTH1R agonists approved to reduce osteoporotic fracture risk in postmenopausal women. This study in 8-week-old female mice compared bone resorption and formation responses to these agents delivered at the same 10 μg/kg dose, and a 40 μg/kg abaloparatide dose was also included to reflect its 4-fold higher approved clinical dose. Peptides or vehicle were administered by daily supra-calvarial subcutaneous injection for 12 days, and local (calvarial) and systemic (L5 vertebral and tibial) responses were evaluated by histomorphometry. Terminal bone histomorphometry data indicated that calvarial resorption cavities were similar in both abaloparatide groups versus vehicle controls, whereas the teriparatide group had more calvarial resorption cavities compared with the vehicle or abaloparatide 40 μg/kg groups. The bone resorption marker serum CTX was significantly lower in the abaloparatide 40 μg/kg group and similar in the other two active treatment groups compared with vehicle controls. Both peptides increased trabecular bone formation rate (BFR) in L5 and proximal tibia versus vehicle, and L5 BFR was higher with abaloparatide 40 μg/kg versus teriparatide. At the tibial diaphysis, periosteal BFR was higher with abaloparatide 40 μg/kg versus vehicle or teriparatide, and endocortical BFR was higher with teriparatide but not with abaloparatide 10 or 40 μg/kg versus vehicle. Few differences in structural or microarchitectural bone parameters were observed with this brief duration of treatment. In summary, calvarial bone resorption cavity counts were higher in the teriparatide group versus the vehicle and abaloparatide 40 μg/kg groups, and the abaloparatide 40 μg/kg group had lower serum CTX versus vehicle. L5 and tibial trabecular bone formation indices were higher in all three active treatment groups versus vehicle. The abaloparatide 40 μg/kg group had higher L5 trabecular BFR and tibial periosteal BFR versus teriparatide, whereas tibial endocortical BFR was higher with teriparatide but not abaloparatide. Together, these findings in female mice indicate that an improved balance of bone formation versus bone resorption is established shortly after initiating treatment with abaloparatide. PTH receptor (PTH-R) agonists increase bone density by stimulating bone formation. PTH-R agonists differ in their propensity to increase bone resorption. Female mice were treated for 12 d with PTH-R agonists abaloparatide or teriparatide. The systemic resorption marker serum CTX was lower with abaloparatide vs vehicle. Calvarial resorption cavities were higher with teriparatide but not abaloparatide.
Collapse
Affiliation(s)
| | | | - Dorothy Hu
- Harvard School of Dental Medicine, Boston, MA, USA
| | - Roland Baron
- Harvard School of Dental Medicine, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | |
Collapse
|
11
|
Wein MN, Kronenberg HM. Regulation of Bone Remodeling by Parathyroid Hormone. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a031237. [PMID: 29358318 DOI: 10.1101/cshperspect.a031237] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Parathyroid hormone (PTH) exerts profound effects on skeletal homeostasis through multiple cellular and molecular mechanisms. Continuous hyperparathyroidism causes net loss of bone mass, despite accelerating bone formation by osteoblasts. Intermittent treatment with PTH analogs represents the only Food and Drug Administration (FDA)-approved bone anabolic osteoporosis treatment strategy. Functional PTH receptors are present on cells of the osteoblast lineage, ranging from early skeletal stem cells to matrix-embedded osteocytes. In addition, bone remodeling by osteoclasts liberates latent growth factors present within bone matrix. Here, we will provide an overview of the multiple cellular and molecular mechanisms through which PTH influences bone homeostasis. Notably, net skeletal effects of continuous versus intermittent can differ significantly. Where possible, we will highlight mechanisms through which continuous hyperparathyroidism leads to bone loss, and through which intermittent hyperparathyroidism boosts bone mass. Given the therapeutic usage of intermittent PTH (iPTH) treatment for osteoporosis, particular attention will be paid toward mechanisms underlying the bone anabolic effects of once daily PTH administration.
Collapse
Affiliation(s)
- Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| | - Henry M Kronenberg
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| |
Collapse
|
12
|
Ebina K, Hirao M, Hashimoto J, Hagihara K, Kashii M, Kitaguchi K, Matsuoka H, Iwahashi T, Chijimatsu R, Yoshikawa H. Assessment of the effects of switching oral bisphosphonates to denosumab or daily teriparatide in patients with rheumatoid arthritis. J Bone Miner Metab 2018; 36:478-487. [PMID: 28766140 DOI: 10.1007/s00774-017-0861-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/12/2017] [Indexed: 01/12/2023]
Abstract
The aim of this observational, non-randomized study was to clarify the unknown effects of switching oral bisphosphonates (BPs) to denosumab (DMAb) or daily teriparatide (TPTD) in patients with rheumatoid arthritis (RA). The characteristics of the 194 female patients included in the study were 183 postmenopausal, age 65.9 years, lumbar spine (LS) T score -1.8, femoral neck (FN) T score -2.3, dose and rate of taking oral prednisolone (3.6 mg/day) 75.8%, and prior BP treatment duration 40.0 months. The patients were allocated to (1) the BP-continue group (n = 80), (2) the switch-to-DMAb group (n = 74), or (3) the switch-to-TPTD group (n = 40). After 18 months, the increase in bone mineral density (BMD) was significantly greater in the switch-to-DMAb group than in the BP-continue group (LS 5.2 vs 2.3%, P < 0.01; FN 3.8 vs 0.0%, P < 0.01) and in the switch-to-TPTD group than in the BP-continue group (LS 9.0 vs 2.3%, P < 0.001; FN 4.9 vs 0.0%, P < 0.01). Moreover, the switch-to-TPTD group showed a higher LS BMD (P < 0.05) and trabecular bone score (TBS) (2.1 vs -0.7%; P < 0.05) increase than the switch-to-DMAb group. Clinical fracture incidence during this period was 8.8% in the BP-continue group, 4.1% in the switch-to-DMAb group, and 2.5% in the switch-to-TPTD group. Both the switch-to-DMAb group and the switch-to-TPTD group showed significant increases in LS and FN BMD, and the switch-to-TPTD group showed a higher increase in TBS compared to the BP-continue group at 18 months. Switching BPs to DMAb or TPTD in female RA may provide some useful osteoporosis treatment options.
Collapse
Affiliation(s)
- Kosuke Ebina
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Makoto Hirao
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Jun Hashimoto
- Department of Rheumatology, National Hospital Organization, Osaka Minami Medical Center, 2-1 Kidohigashi, Kawachinagano, Osaka, 586-8521, Japan
| | - Keisuke Hagihara
- Department of Kampo Medicine, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masafumi Kashii
- Department of Orthopaedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibahara, Toyonaka, Osaka, 560-8565, Japan
| | - Kazuma Kitaguchi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hozo Matsuoka
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toru Iwahashi
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryota Chijimatsu
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
13
|
Di Nisio A, De Toni L, Rocca MS, Ghezzi M, Selice R, Taglialavoro G, Ferlin A, Foresta C. Negative Association Between Sclerostin and INSL3 in Isolated Human Osteocytes and in Klinefelter Syndrome: New Hints for Testis-Bone Crosstalk. J Clin Endocrinol Metab 2018; 103:2033-2041. [PMID: 29452406 DOI: 10.1210/jc.2017-02762] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 02/09/2018] [Indexed: 11/19/2022]
Abstract
CONTEXT The regulation of bone mass by the testis is a well-recognized mechanism, but the role of Leydig-specific marker insulin-like 3 peptide (INSL3) on the most abundant bone cell population, osteocytes, is unknown. In this study, we aimed to investigate the relationship between INSL3 and sclerostin, an osteocyte-specific protein that negatively regulates bone formation. DESIGN Serum sclerostin and INSL3 levels were evaluated in Klinefelter syndrome (KS) and healthy controls. In vitro effect of INSL3 on sclerostin production was evaluated in human cultured osteocytes. PATIENTS A total of 103 KS patients and 60 age- and sex-matched controls were recruited. MAIN OUTCOME MEASURES Serum sclerostin and INSL3 levels were assessed by enzyme-linked immunosorbent assay. Osteocytes were isolated by fluorescence-assisted cell sorting. Sclerostin expression was evaluated by western blot, immunofluorescence, and reverse transcription polymerase chain reaction. Measurement of bone mineral density was done by dual-energy X-ray absorptiometry at lumbar spine (L1-L4) and femoral neck. RESULTS Sclerostin levels were significantly increased in KS subjects, and negatively correlated with INSL3 levels in both cohorts and with bone mineral density in the KS group. Stimulation of cultured osteocytes with INSL3 at 10-7 M significantly decreased both sclerostin messenger RNA and protein expression. CONCLUSIONS We report a negative association between the testicular hormone INSL3 and the osteocytic negative regulator of bone formation, sclerostin. We further explored this association in vitro and showed that INSL3 was able to reduce sclerostin expression. These results add further knowledge on the emerging role of sclerostin as a therapeutic target for osteoporosis treatment.
Collapse
Affiliation(s)
- Andrea Di Nisio
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Padova, Italy
| | - Luca De Toni
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Padova, Italy
| | - Maria Santa Rocca
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Padova, Italy
| | - Marco Ghezzi
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Padova, Italy
| | - Riccardo Selice
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Padova, Italy
| | - Giuseppe Taglialavoro
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy
| | - Alberto Ferlin
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Padova, Italy
| | - Carlo Foresta
- Department of Medicine, Operative Unit of Andrology and Medicine of Human Reproduction, University of Padova, Padova, Italy
| |
Collapse
|
14
|
Wein MN. Parathyroid Hormone Signaling in Osteocytes. JBMR Plus 2017; 2:22-30. [PMID: 30283888 PMCID: PMC6124166 DOI: 10.1002/jbm4.10021] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/10/2017] [Accepted: 10/12/2017] [Indexed: 12/13/2022] Open
Abstract
Osteocytes are the most abundant cell type in bone and play a central role in orchestrating skeletal remodeling, in part by producing paracrine‐acting factors that in turn influence osteoblast and osteoclast activity. Recent evidence has indicated that osteocytes are crucial cellular targets of parathyroid hormone (PTH). Here, we will review the cellular and molecular mechanisms through which PTH influences osteocyte function. Two well‐studied PTH target genes in osteocytes are SOST and receptor activator of NF‐κB ligand (RANKL). The molecular mechanisms through which PTH regulates expression of these two crucial target genes will be discussed. Beyond SOST and RANKL, PTH/PTH‐related peptide (PTHrP) signaling in osteocytes may directly influence the way osteocytes remodel their perilacunar environment to influence bone homeostasis in a cell‐autonomous manner. Here, I will highlight novel, additional mechanisms used by PTH and PTHrP to modulate bone homeostasis through effects in osteocytes. © 2017 The Authors. JBMR Plus is published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Marc N Wein
- Endocrine Unit, Massachusetts General Hospital Harvard Medical School Boston MA USA
| |
Collapse
|
15
|
Siddiqui JA, Partridge NC. Physiological Bone Remodeling: Systemic Regulation and Growth Factor Involvement. Physiology (Bethesda) 2017; 31:233-45. [PMID: 27053737 DOI: 10.1152/physiol.00061.2014] [Citation(s) in RCA: 252] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Bone remodeling is essential for adult bone homeostasis. It comprises two phases: bone formation and resorption. The balance between the two phases is crucial for sustaining bone mass and systemic mineral homeostasis. This review highlights recent work on physiological bone remodeling and discusses our knowledge of how systemic and growth factors regulate this process.
Collapse
Affiliation(s)
- Jawed A Siddiqui
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - Nicola C Partridge
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| |
Collapse
|
16
|
Delgado-Calle J, Tu X, Pacheco-Costa R, McAndrews K, Edwards R, Pellegrini GG, Kuhlenschmidt K, Olivos N, Robling A, Peacock M, Plotkin LI, Bellido T. Control of Bone Anabolism in Response to Mechanical Loading and PTH by Distinct Mechanisms Downstream of the PTH Receptor. J Bone Miner Res 2017; 32:522-535. [PMID: 27704638 PMCID: PMC8502039 DOI: 10.1002/jbmr.3011] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/30/2016] [Accepted: 10/04/2016] [Indexed: 12/31/2022]
Abstract
Osteocytes integrate the responses of bone to mechanical and hormonal stimuli by poorly understood mechanisms. We report here that mice with conditional deletion of the parathyroid hormone (PTH) receptor 1 (Pth1r) in dentin matrix protein 1 (DMP1)-8kb-expressing cells (cKO) exhibit a modest decrease in bone resorption leading to a mild increase in cancellous bone without changes in cortical bone. However, bone resorption in response to endogenous chronic elevation of PTH in growing or adult cKO mice induced by a low calcium diet remained intact, because the increased bone remodeling and bone loss was indistinguishable from that exhibited by control littermates. In contrast, the bone gain and increased bone formation in cancellous and cortical bone induced by daily injections of PTH and the periosteal bone apposition induced by axial ulna loading were markedly reduced in cKO mice compared to controls. Remarkably, however, wild-type (WT) control littermates and transgenic mice overexpressing SOST injected daily with PTH exhibit similar activation of Wnt/β-catenin signaling, increased bone formation, and cancellous and cortical bone gain. Taken together, these findings demonstrate that Pth1r in DMP1-8kb-expressing cells is required to maintain basal levels of bone resorption but is dispensable for the catabolic action of chronic PTH elevation; and it is essential for the anabolic actions of daily PTH injections and mechanical loading. However, downregulation of Sost/sclerostin, previously shown to be required for bone anabolism induced by mechanical loading, is not required for PTH-induced bone gain, showing that other mechanisms downstream of the Pth1r in DMP1-8kb-expressing cells are responsible for the hormonal effect. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jesus Delgado-Calle
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Xiaolin Tu
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Rafael Pacheco-Costa
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Kevin McAndrews
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Rachel Edwards
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Gretel G Pellegrini
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Kali Kuhlenschmidt
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Naomie Olivos
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Alexander Robling
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.,Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA
| | - Munro Peacock
- Department of Medicine, Division of Endocrinology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Lilian I Plotkin
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, IN, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.,Department of Medicine, Division of Endocrinology, School of Medicine, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
17
|
Delgado-Calle J, Sato AY, Bellido T. Role and mechanism of action of sclerostin in bone. Bone 2017; 96:29-37. [PMID: 27742498 PMCID: PMC5328835 DOI: 10.1016/j.bone.2016.10.007] [Citation(s) in RCA: 286] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/29/2016] [Accepted: 10/10/2016] [Indexed: 12/14/2022]
Abstract
After discovering that lack of Sost/sclerostin expression is the cause of the high bone mass human syndromes Van Buchem disease and sclerosteosis, extensive animal experimentation and clinical studies demonstrated that sclerostin plays a critical role in bone homeostasis and that its deficiency or pharmacological neutralization increases bone formation. Dysregulation of sclerostin expression also underlies the pathophysiology of skeletal disorders characterized by loss of bone mass, as well as the damaging effects of some cancers in bone. Thus, sclerostin has quickly become a promising molecular target for the treatment of osteoporosis and other skeletal diseases, and beneficial skeletal outcomes are observed in animal studies and clinical trials using neutralizing antibodies against sclerostin. However, the anabolic effect of blocking sclerostin decreases with time, bone mass accrual is also accompanied by anti-catabolic effects, and there is bone loss over time after therapy discontinuation. Further, the cellular source of sclerostin in the bone/bone marrow microenvironment under physiological and pathological conditions, the pathways that regulate sclerostin expression and the mechanisms by which sclerostin modulates the activity of osteocytes, osteoblasts, and osteoclasts remain unclear. In this review, we highlight the current knowledge on the regulation of Sost/sclerotin expression and its mechanism(s) of action, discuss novel observations regarding its role in signaling pathways activated by hormones and mechanical stimuli in bone, and propose future research needed to understand the full potential of therapeutic interventions that modulate Sost/sclerostin expression.
Collapse
Affiliation(s)
- Jesus Delgado-Calle
- Department of Anatomy and Cell Biology, Indianapolis, IN, United States; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States.
| | - Amy Y Sato
- Department of Anatomy and Cell Biology, Indianapolis, IN, United States.
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indianapolis, IN, United States; Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, United States; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States.
| |
Collapse
|
18
|
Osagie-Clouard L, Sanghani A, Coathup M, Briggs T, Bostrom M, Blunn G. Parathyroid hormone 1-34 and skeletal anabolic action: The use of parathyroid hormone in bone formation. Bone Joint Res 2017; 6:14-21. [PMID: 28062525 PMCID: PMC5227055 DOI: 10.1302/2046-3758.61.bjr-2016-0085.r1] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 10/24/2016] [Indexed: 12/19/2022] Open
Abstract
Intermittently administered parathyroid hormone (PTH 1-34) has been shown to promote bone formation in both human and animal studies. The hormone and its analogues stimulate both bone formation and resorption, and as such at low doses are now in clinical use for the treatment of severe osteoporosis. By varying the duration of exposure, parathyroid hormone can modulate genes leading to increased bone formation within a so-called 'anabolic window'. The osteogenic mechanisms involved are multiple, affecting the stimulation of osteoprogenitor cells, osteoblasts, osteocytes and the stem cell niche, and ultimately leading to increased osteoblast activation, reduced osteoblast apoptosis, upregulation of Wnt/β-catenin signalling, increased stem cell mobilisation, and mediation of the RANKL/OPG pathway. Ongoing investigation into their effect on bone formation through 'coupled' and 'uncoupled' mechanisms further underlines the impact of intermittent PTH on both cortical and cancellous bone. Given the principally catabolic actions of continuous PTH, this article reviews the skeletal actions of intermittent PTH 1-34 and the mechanisms underlying its effect. CITE THIS ARTICLE L. Osagie-Clouard, A. Sanghani, M. Coathup, T. Briggs, M. Bostrom, G. Blunn. Parathyroid hormone 1-34 and skeletal anabolic action: The use of parathyroid hormone in bone formation. Bone Joint Res 2017;6:14-21. DOI: 10.1302/2046-3758.61.BJR-2016-0085.R1.
Collapse
Affiliation(s)
- L Osagie-Clouard
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - A Sanghani
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - M Coathup
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - T Briggs
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - M Bostrom
- Hospital for Special Surgery, New York, New York, USA
| | - G Blunn
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| |
Collapse
|
19
|
Abstract
Osteocytes are differentiated osteoblasts that become surrounded by matrix during the process of bone formation. Acquisition of the osteocyte phenotype is achieved by profound changes in gene expression that facilitate adaptation to the changing cellular environment and constitute the molecular signature of osteocytes. During osteocytogenesis, the expression of genes that are characteristic of the osteoblast are altered and the expression of genes and/or proteins that impart dendritic cellular morphology, regulate matrix mineralization and control the function of cells at the bone surface are ordely modulated. The discovery of mutations in human osteocytic genes has contributed, in a large part, to our understanding of the role of osteocytes in bone homeostasis. Osteocytes are targets of the mechanical force imposed on the skeleton and have a critical role in integrating mechanosensory pathways with the action of hormones, which thereby leads to the orchestrated response of bone to environmental cues. Current, therapeutic approaches harness this accumulating knowledge by targeting osteocytic signalling pathways and messengers to improve skeletal health.
Collapse
Affiliation(s)
- Lilian I. Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine
- Roudebush Veterans Administration Medical Center, Indianapolis, IN
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine
- Roudebush Veterans Administration Medical Center, Indianapolis, IN
| |
Collapse
|
20
|
Leder BZ, Tsai JN, Burnett-Bowie SA, Bouxsein ML, Neer RM. Letter to the editor in response to the commentary, "Concurrent administration of PTH and antiresorptives: Additive effects or DXA cosmetics. Bone 2016; 89:73-74. [PMID: 27157641 DOI: 10.1016/j.bone.2016.04.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 04/25/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Benjamin Z Leder
- Department of Medicine, Endocrine Unit, Massachusetts General Hospital, Boston, MA, United States.
| | - Joy N Tsai
- Department of Medicine, Endocrine Unit, Massachusetts General Hospital, Boston, MA, United States
| | | | - Mary L Bouxsein
- Department of Medicine, Endocrine Unit, Massachusetts General Hospital, Boston, MA, United States; Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Robert M Neer
- Department of Medicine, Endocrine Unit, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
21
|
Di Nisio A, De Toni L, Speltra E, Rocca MS, Taglialavoro G, Ferlin A, Foresta C. Regulation of Sclerostin Production in Human Male Osteocytes by Androgens: Experimental and Clinical Evidence. Endocrinology 2015; 156:4534-44. [PMID: 26393301 DOI: 10.1210/en.2015-1244] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this study we aimed to elucidate a possible role of T in the regulation of sclerostin, a glycoprotein secreted by osteocytes known to regulate bone mass. To this end, we evaluated the effect of T stimulation on sclerostin production and gene expression in human cultured osteocytes. In addition, we evaluated serum sclerostin levels in a cohort of 20 hypogonadal male patients, compared with 20 age-matched eugonadal controls. Stimulation with DHT decreased sclerostin expression in cultured osteocytes in a time- and dose-dependent manner. Confirming a direct androgen receptor-mediated effect on sclerostin production, flutamide coincubation and silencing of androgen receptor gene in osteocytes abolished the DHT effects. In addition, hypogonadal patients showed higher serum sclerostin levels with respect to controls (145.87 ± 50.83 pg/mL vs 84.02 ± 32.15 pg/mL; P < .001) and in both probands and controls, serum T levels were negatively correlated with sclerostin (R = -0.664, P = 0.007, and R = -0.447, P = .045, respectively). Finally, multiple stepwise regression analysis showed that T represented the only independent predictor of sclerostin levels. In conclusion, by showing a direct correlation between T and sclerostin, both in vivo and in vitro, this study adds further support to the emerging clinical and experimental studies focusing on sclerostin as a therapeutic target for osteoporosis treatment.
Collapse
Affiliation(s)
- Andrea Di Nisio
- Department of Medicine (A.D.N., L.D.T., E.S., M.S.R., A.F., C.F.), Operative Unit of Andrology and Medicine of Human Reproduction, and Department of Surgical, Oncological, and Gastroenterological Sciences (G.T.), University of Padova, 35128 Padova, Italy
| | - Luca De Toni
- Department of Medicine (A.D.N., L.D.T., E.S., M.S.R., A.F., C.F.), Operative Unit of Andrology and Medicine of Human Reproduction, and Department of Surgical, Oncological, and Gastroenterological Sciences (G.T.), University of Padova, 35128 Padova, Italy
| | - Elena Speltra
- Department of Medicine (A.D.N., L.D.T., E.S., M.S.R., A.F., C.F.), Operative Unit of Andrology and Medicine of Human Reproduction, and Department of Surgical, Oncological, and Gastroenterological Sciences (G.T.), University of Padova, 35128 Padova, Italy
| | - Maria Santa Rocca
- Department of Medicine (A.D.N., L.D.T., E.S., M.S.R., A.F., C.F.), Operative Unit of Andrology and Medicine of Human Reproduction, and Department of Surgical, Oncological, and Gastroenterological Sciences (G.T.), University of Padova, 35128 Padova, Italy
| | - Giuseppe Taglialavoro
- Department of Medicine (A.D.N., L.D.T., E.S., M.S.R., A.F., C.F.), Operative Unit of Andrology and Medicine of Human Reproduction, and Department of Surgical, Oncological, and Gastroenterological Sciences (G.T.), University of Padova, 35128 Padova, Italy
| | - Alberto Ferlin
- Department of Medicine (A.D.N., L.D.T., E.S., M.S.R., A.F., C.F.), Operative Unit of Andrology and Medicine of Human Reproduction, and Department of Surgical, Oncological, and Gastroenterological Sciences (G.T.), University of Padova, 35128 Padova, Italy
| | - Carlo Foresta
- Department of Medicine (A.D.N., L.D.T., E.S., M.S.R., A.F., C.F.), Operative Unit of Andrology and Medicine of Human Reproduction, and Department of Surgical, Oncological, and Gastroenterological Sciences (G.T.), University of Padova, 35128 Padova, Italy
| |
Collapse
|
22
|
Niedźwiedzki T, Filipowska J. Bone remodeling in the context of cellular and systemic regulation: the role of osteocytes and the nervous system. J Mol Endocrinol 2015; 55:R23-36. [PMID: 26307562 DOI: 10.1530/jme-15-0067] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/24/2015] [Indexed: 12/30/2022]
Abstract
Bone is a dynamic tissue that undergoes constant remodeling. The appropriate course of this process determines development and regeneration of the skeleton. Tight molecular control of bone remodeling is vital for the maintenance of appropriate physiology and microarchitecture of the bone, providing homeostasis, also at the systemic level. The process of remodeling is regulated by a rich innervation of the skeleton, being the source of various growth factors, neurotransmitters, and hormones regulating function of the bone. Although the course of bone remodeling at the cellular level is mainly associated with the activity of osteoclasts and osteoblasts, recently also osteocytes have gained a growing interest as the principal regulators of bone turnover. Osteocytes play a significant role in the regulation of osteogenesis, releasing sclerostin (SOST), an inhibitor of bone formation. The process of bone turnover, especially osteogenesis, is also modulated by extra-skeletal molecules. Proliferation and differentiation of osteoblasts are promoted by the brain-derived serotonin and hypothetically inhibited by its intestinal equivalent. The activity of SOST and serotonin is either directly or indirectly associated with the canonical Wnt/β-catenin signaling pathway, the main regulatory pathway of osteoblasts function. The impairment of bone remodeling may lead to many skeletal diseases, such as high bone mass syndrome or osteoporosis. In this paper, we review the most recent data on the cellular and molecular mechanisms of bone remodeling control, with particular emphasis on the role of osteocytes and the nervous system in this process.
Collapse
Affiliation(s)
- Tadeusz Niedźwiedzki
- Department of Orthopedics and PhysiotherapyCollegium Medicum, Jagiellonian University, Cracow, PolandDepartment of Cell Biology and ImagingInstitute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Cracow, Poland
| | - Joanna Filipowska
- Department of Orthopedics and PhysiotherapyCollegium Medicum, Jagiellonian University, Cracow, PolandDepartment of Cell Biology and ImagingInstitute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Cracow, Poland
| |
Collapse
|
23
|
Babey M, Wang Y, Kubota T, Fong C, Menendez A, ElAlieh HZ, Bikle DD. Gender-Specific Differences in the Skeletal Response to Continuous PTH in Mice Lacking the IGF1 Receptor in Mature Osteoblasts. J Bone Miner Res 2015; 30:1064-76. [PMID: 25502173 PMCID: PMC9045460 DOI: 10.1002/jbmr.2433] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/28/2014] [Accepted: 12/10/2014] [Indexed: 12/26/2022]
Abstract
The primary goal of this study was to determine whether the IGF1R in mature osteoblasts and osteocytes was required for the catabolic actions of continuous parathyroid hormone (cPTH). Igf1r was deleted from male and female FVN/B mice by breeding with mice expressing cre recombinase under control of the osteocalcin promoter ((0CN) Igfr1(-/-) ). Littermates lacking the cre recombinase served as controls. PTH, 60 μg/kg/d, was administered continuously by Alzet minipumps for 4 weeks. Blood was obtained for indices of calcium metabolism. The femurs were examined by micro-computed tomography for structure, immunohistochemistry for IGF1R expression, histomorphometry for bone formation rates (BFR), mRNA levels by qPCR, and bone marrow stromal cell cultures (BMSC) for alkaline phosphatase activity (ALP(+) ), mineralization, and osteoblast-induced osteoclastogenesis. Whereas cPTH led to a reduction in trabecular bone volume/tissue volume (BV/TV) and cortical thickness in the control females, no change was found in the control males. Although trabecular BV/TV and cortical thickness were reduced in the (0CN) Igfr1(-/-) mice of both sexes, no further reduction after cPTH was found in the females, unlike the reduction in males. BFR was stimulated by cPTH in the controls but blocked by Igf1r deletion in the females. The (0CN) Igfr1(-/-) male mice showed a partial response. ALP(+) and mineralized colony formation were higher in BMSC from control males than from control females. These markers were increased by cPTH in both sexes, but BMSC from male (0CN) Igfr1(-/-) also were increased by cPTH, unlike those from female (0CN) Igfr1(-/-) . cPTH stimulated receptor activator of NF-κB ligand (RANKL) and decreased osteoprotegerin and alkaline phosphatase expression more in control female bone than in control male bone. Deletion of Igf1r blocked these effects of cPTH in the female but not in the male. However, PTH stimulation of osteoblast-driven osteoclastogenesis was blocked by deleting Igfr1 in both sexes. We conclude that cPTH is catabolic in female but not male mice. Moreover, IGF1 signaling plays a greater role in the skeletal actions of cPTH in the female mouse than in the male mouse, which may underlie the sex differences in the response to cPTH.
Collapse
Affiliation(s)
- Muriel Babey
- Endocrine Research Unit, University of California, San Francisco, CA, USA
| | - Yongmei Wang
- Endocrine Research Unit, University of California, San Francisco, CA, USA
| | - Takuo Kubota
- Endocrine Research Unit, University of California, San Francisco, CA, USA
| | - Chak Fong
- Endocrine Research Unit, University of California, San Francisco, CA, USA
| | - Alicia Menendez
- Endocrine Research Unit, University of California, San Francisco, CA, USA
| | - Hashem Z ElAlieh
- Endocrine Research Unit, University of California, San Francisco, CA, USA
| | - Daniel D Bikle
- Endocrine Research Unit, University of California, San Francisco, CA, USA
| |
Collapse
|
24
|
Gaudio A, Muratore F, Fiore V, Rapisarda R, Signorelli SS, Fiore CE. Decreased bone cortical density at the forearm in subjects with subclinical peripheral arterial disease. Osteoporos Int 2015; 26:1747-53. [PMID: 25672808 DOI: 10.1007/s00198-015-3057-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/28/2015] [Indexed: 12/14/2022]
Abstract
UNLABELLED The association between peripheral arterial disease (PAD) and low bone mass is controversial. In our study, peripheral quantitative computed tomography shows a reduction of cortical but not trabecular, bone mineral density (BMD) at the forearm, in patients with subclinical PAD. INTRODUCTION Some controversy exists regarding the association between peripheral arterial disease (PAD) and low bone mass. Previous studies have evaluated bone mineral density (BMD) in patients with subclinical PAD, with mixed results. Inconsistency of data may depend on the fact that most studies measured areal bone mineral density (aBMD) by Dual-energy-x ray absorptiometry (DXA). Because DXA cannot distinguish between cortical and trabecular compartments, we reasoned that a study aimed to establish whether these compartments were differentially affected by PAD status could give more information on the nature of this association. METHODS In this cross-sectional study, we used peripheral quantitative computed tomography (pQCT) to examine volumetric cortical and trabecular mineral density at the radius (vBMD) in a cohort of subjects with subclinical PAD as defined by ABI ≤0.90 and compared them with healthy subjects with no evidence of PAD. RESULTS Patients with subclinical PAD had significantly reduced cortical density (1101.0 ± 45.4 vs 1156.2 ± 51.3 mg/cm(3), p < 0.001) and cortical area (75.0 ± 20.9 vs 99.9 ± 18.2 mm(2), p < 0.001) than healthy subjects. Trabecular density (178.1 ± 47.9 vs 165.8 ± 29.6 mg/cm(3)) was not significantly different in the two groups. CONCLUSION Subclinical PAD induces a selective bone loss at the radius compartment, not identified by standard DXA, which seems to occur primarily at the cortical level.
Collapse
Affiliation(s)
- A Gaudio
- Department of Clinical and Experimental Medicine, Section of Metabolic Bone Disease, University of Catania, Clinica Medica OVE, Via Plebiscito 628, 95124, Catania, Italy
| | - F Muratore
- Department of Clinical and Experimental Medicine, Section of Metabolic Bone Disease, University of Catania, Clinica Medica OVE, Via Plebiscito 628, 95124, Catania, Italy
| | - V Fiore
- Department of Clinical and Experimental Medicine, Section of Vascular Medicine, Medical Angiology Unit, University of Catania, Garibaldi Hospital, Piazza S. Maria di Gesù 5, 95123, Catania, Italy
| | - R Rapisarda
- Department of Clinical and Experimental Medicine, Section of Metabolic Bone Disease, University of Catania, Clinica Medica OVE, Via Plebiscito 628, 95124, Catania, Italy
| | - S S Signorelli
- Department of Clinical and Experimental Medicine, Section of Vascular Medicine, Medical Angiology Unit, University of Catania, Garibaldi Hospital, Piazza S. Maria di Gesù 5, 95123, Catania, Italy
| | - C E Fiore
- Department of Clinical and Experimental Medicine, Section of Metabolic Bone Disease, University of Catania, Clinica Medica OVE, Via Plebiscito 628, 95124, Catania, Italy.
| |
Collapse
|
25
|
Silva BC, Bilezikian JP. Parathyroid hormone: anabolic and catabolic actions on the skeleton. Curr Opin Pharmacol 2015; 22:41-50. [PMID: 25854704 DOI: 10.1016/j.coph.2015.03.005] [Citation(s) in RCA: 322] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/24/2015] [Indexed: 12/16/2022]
Abstract
Parathyroid hormone (PTH) is essential for the maintenance of calcium homeostasis through, in part, its actions to regulate bone remodeling. While PTH stimulates both bone formation and bone resorption, the duration and periodicity of exposure to PTH governs the net effect on bone mass, that is whether it is catabolic or anabolic. PTH receptor signaling in osteoblasts and osteocytes can increase the RANKL/OPG ratio, increasing both osteoclast recruitment and osteoclast activity, and thereby stimulating bone resorption. In contrast, PTH-induced bone formation is explained, at least in part, by its ability to downregulate SOST/sclerostin expression in osteocytes, permitting the anabolic Wnt signaling pathway to proceed. The two modes of administration of PTH, that is, continuous vs. intermittent, can regulate, in bone cells, different sets of genes; alternatively, the same sets of genes exposed to PTH in sustained vs. transient way, will favor bone resorption or bone formation, respectively. This article reviews the effects of PTH on bone cells that lead to these dual catabolic and anabolic actions on the skeleton.
Collapse
Affiliation(s)
- Barbara C Silva
- Santa Casa de Belo Horizonte and Felicio Rocho Hospital, Division of Endocrinology, Brazil
| | - John P Bilezikian
- Metabolic Bone Diseases Unit, Division of Endocrinology, Department of Medicine, College of Physicians and Surgeons, Columbia University, United States.
| |
Collapse
|
26
|
Moe SM, Chen NX, Newman CL, Organ JM, Kneissel M, Kramer I, Gattone VH, Allen MR. Anti-sclerostin antibody treatment in a rat model of progressive renal osteodystrophy. J Bone Miner Res 2015; 30:499-509. [PMID: 25407607 PMCID: PMC4333005 DOI: 10.1002/jbmr.2372] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 09/15/2014] [Accepted: 09/20/2014] [Indexed: 12/20/2022]
Abstract
Chronic kidney disease (CKD) is associated with abnormalities in bone quantity and quality, leading to increased fractures. Recent studies suggest abnormalities of Wnt signaling in animal models of CKD and elevated sclerostin levels in patients with CKD. The goal of this study was to evaluate the effectiveness of anti-sclerostin antibody treatment in an animal model of progressive CKD with low and high parathyroid hormone (PTH) levels. Cy/+ male rats (CKD) were treated without or with calcium in the drinking water at 25 weeks of age to stratify the animals into high PTH and low PTH groups, respectively, by 30 weeks. Animals were then treated with anti-sclerostin antibody at 100 mg/kg i.v. weekly for 5 doses, a single 20-µg/kg subcutaneous dose of zoledronic acid, or no treatment, and were then euthanized at 35 weeks. As a positive control, the efficacy of anti-sclerostin antibody treatment was also evaluated in normal littermates. The results demonstrated that the CKD animals with high PTH had lower calcium, higher phosphorus, and lower FGF23 compared to the CKD animals with low PTH. Treatment with anti-sclerostin antibody had no effect on any of the biochemistries, whereas zoledronic acid lowered dkk-1 levels. The anti-sclerostin antibody increased trabecular bone volume/total volume (BV/TV) and trabecular mineralization surface in animals with low PTH, but not in animals with high PTH. Neither anti-sclerostin antibody nor zoledronic acid improved biomechanical properties in the animals. Cortical porosity was severe in high-PTH animals and was unaffected by either treatment. In contrast, in normal animals treated with anti-sclerostin antibody, there was an improvement in bone volume, cortical geometry, and biomechanical properties. In summary, this is the first study to test the efficacy of anti-sclerostin antibody treatment on animals with advanced CKD. We found efficacy in improving bone properties only when the PTH levels were low.
Collapse
Affiliation(s)
- Sharon M Moe
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Roduebush Veterans Affairs Medical Center, Indianapolis, IN, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Osteocyte-derived insulin-like growth factor I is not essential for the bone repletion response in mice. PLoS One 2015; 10:e0115897. [PMID: 25635763 PMCID: PMC4312049 DOI: 10.1371/journal.pone.0115897] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 11/28/2014] [Indexed: 12/17/2022] Open
Abstract
The present study sought to evaluate the functional role of osteocyte-derived IGF-I in the bone repletion process by determining whether deficient expression of Igf1 in osteocytes would impair the bone repletion response to one week of dietary calcium repletion after two weeks of dietary calcium deprivation. As expected, the two-week dietary calcium depletion led to hypocalcemia, secondary hyperparathyroidism, and increases in bone resorption and bone loss in both Igf1 osteocyte conditional knockout (cKO) mutants and WT control mice. Thus, conditional disruption of Igf1 in osteocytes did not impair the calcium depletion-induced bone resorption. After one week of calcium repletion, both cKO mutants and WT littermates showed an increase in endosteal bone formation attended by the reduction in osteoclast number, indicating that deficient Igf1 expression in osteocytes also did not have deleterious effects on the bone repletion response. The lack of an effect of deficient osteocyte-derived IGF-I expression on bone repletion is unexpected since previous studies show that these Igf1 osteocyte cKO mice exhibited impaired developmental growth and displayed complete resistance to bone anabolic effects of loading. These studies suggest that there is a dichotomy between the mechanisms necessary for anabolic responses to mechanical loading and the regulatory hormonal and anabolic skeletal repletion following low dietary calcium challenge. In conclusion, to our knowledge this study has demonstrated for the first time that osteocyte-derived IGF-I, which is essential for anabolic bone response to mechanical loading, is not a key regulatory factor for bone repletion after a low calcium challenge.
Collapse
|
28
|
Osteocytes mediate the anabolic actions of canonical Wnt/β-catenin signaling in bone. Proc Natl Acad Sci U S A 2015; 112:E478-86. [PMID: 25605937 DOI: 10.1073/pnas.1409857112] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Osteocytes, >90% of the cells in bone, lie embedded within the mineralized matrix and coordinate osteoclast and osteoblast activity on bone surfaces by mechanisms still unclear. Bone anabolic stimuli activate Wnt signaling, and human mutations of components along this pathway underscore its crucial role in bone accrual and maintenance. However, the cell responsible for orchestrating Wnt anabolic actions has remained elusive. We show herein that activation of canonical Wnt signaling exclusively in osteocytes [dominant active (da)βcat(Ot) mice] induces bone anabolism and triggers Notch signaling without affecting survival. These features contrast with those of mice expressing the same daß-catenin in osteoblasts, which exhibit decreased resorption and perinatal death from leukemia. daßcat(Ot) mice exhibit increased bone mineral density in the axial and appendicular skeleton, and marked increase in bone volume in cancellous/trabecular and cortical compartments compared with littermate controls. daßcat(Ot) mice display increased resorption and formation markers, high number of osteoclasts and osteoblasts in cancellous and cortical bone, increased bone matrix production, and markedly elevated periosteal bone formation rate. Wnt and Notch signaling target genes, osteoblast and osteocyte markers, and proosteoclastogenic and antiosteoclastogenic cytokines are elevated in bones of daßcat(Ot) mice. Further, the increase in RANKL depends on Sost/sclerostin. Thus, activation of osteocytic β-catenin signaling increases both osteoclasts and osteoblasts, leading to bone gain, and is sufficient to activate the Notch pathway. These findings demonstrate disparate outcomes of β-catenin activation in osteocytes versus osteoblasts and identify osteocytes as central target cells of the anabolic actions of canonical Wnt/β-catenin signaling in bone.
Collapse
|
29
|
Abstract
Fractures across the stages of chronic kidney disease (CKD) could be due to osteoporosis, some form of renal osteodystrophy defined by specific quantitative histomorphometry or chronic kidney disease-mineral and bone disorder (CKD-MBD). CKD-MBD is a systemic disease that links disorders of mineral and bone metabolism due to CKD to either one or all of the following: abnormalities of calcium, phosphorus, parathyroid hormone or vitamin D metabolism; abnormalities in bone turnover, mineralization, volume, linear growth or strength; or vascular or other soft-tissue calcification. Osteoporosis, as defined by the National Institutes of Health, may coexist with renal osteodystrophy or CKD-MBD. Differentiation among these disorders is required to manage correctly the correct disorder to reduce the risk of fractures. While the World Health Organization (WHO) bone mineral density (BMD) criteria for osteoporosis can be used in patients with stages 1-3 CKD, the disorders of bone turnover become so aberrant by stages 4 and 5 CKD that neither the WHO criteria nor the occurrence of a fragility fracture can be used for the diagnosis of osteoporosis. The diagnosis of osteoporosis in stages 4 and 5 CKD is one of the exclusion-excluding either renal osteodystrophy or CKD-MBD as the cause of low BMD or fragility fractures. Differentiations among the disorders of renal osteodystrophy, CKD-MBD or osteoporosis are dependent on the measurement of specific biochemical markers, including serum parathyroid hormone (PTH) and/or quantitative bone histomorphometry. Management of fractures in stages 1-3 CKD does not differ in persons with or without CKD with osteoporosis assuming that there is no evidence for CKD-MBD, clinically suspected by elevated PTH, hyperphosphatemia or fibroblast growth factor 23 due to CKD. Treatment of fractures in persons with osteoporosis and stages 4 and 5 CKD is not evidence-based, with the exception of post-hoc analysis suggesting efficacy and safety of specific osteoporosis therapies (alendronate, risedronate and denosumab) in stage 4 CKD. This review also discusses how to diagnose and manage fragility fractures across the five stages of CKD.
Collapse
Affiliation(s)
- Paul D Miller
- Colorado Center for Bone Research, Lakewood, CO, USA
| |
Collapse
|
30
|
Ebina K, Hashimoto J, Shi K, Kashii M, Hirao M, Yoshikawa H. Comparison of the effect of 18-month daily teriparatide administration on patients with rheumatoid arthritis and postmenopausal osteoporosis patients. Osteoporos Int 2014; 25:2755-65. [PMID: 25082556 DOI: 10.1007/s00198-014-2819-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/22/2014] [Indexed: 01/09/2023]
Abstract
UNLABELLED Patients with rheumatoid arthritis showed greater response to 18-month administration of daily teriparatide especially in the increase of bone formation markers at 1 month and femoral neck bone mineral density at 18 months compared to postmenopausal osteoporosis patients. INTRODUCTION The aim of this study was to evaluate the effects of 18-month administration of daily teriparatide (TPTD) in osteoporosis patients with rheumatoid arthritis (RA) by comparing that of postmenopausal osteoporosis patients (Porosis). METHODS The effects of TPTD were examined between RA (n = 70; age 68.4 years; disease activity score assessing 28 joints with CRP [DAS28-CRP] 2.8; rheumatoid factor [RF] positivity 75.5 %) with 77.1 % of prior bisphosphonate (BP), 84.3 % of oral prednisolone (PSL) (4.4 mg/day at baseline), 25.7 % of biologics, and Porosis (n = 62; age 71.3 years) with 77.4 % of prior BP. RESULTS Femoral neck (FN) bone mineral density (BMD) increase at 18 months was significantly greater in RA compared to Porosis (4.7 vs. 0.7 %, P = 0.038), whereas it was 9.7 versus 7.9 % (P = 0.736) in the lumbar spine (LS). The increase of bone formation markers (bone alkaline phosphatase [bone ALP] and N-terminal type I procollagen propeptide [PINP]) at 1 month were all significantly greater in RA compared to Porosis. A multivariate logistic regression analysis revealed that the significant indicator of 18-month BMD increase in RA was a 3-month increase of under-carboxylated osteocalcin (ucOC) for LS (β = 0.446, P = 0.005) and baseline ucOC for FN (β = 0.554, P = 0.001), in which both showed significant negative correlation with baseline PSL dose. CONCLUSIONS RA showed greater response to daily TPTD administration, especially in the increase of bone formation markers at 1 month and FN BMD increase at 18 months compared to Porosis.
Collapse
Affiliation(s)
- K Ebina
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan,
| | | | | | | | | | | |
Collapse
|
31
|
Ben-awadh AN, Delgado-Calle J, Tu X, Kuhlenschmidt K, Allen MR, Plotkin LI, Bellido T. Parathyroid hormone receptor signaling induces bone resorption in the adult skeleton by directly regulating the RANKL gene in osteocytes. Endocrinology 2014; 155:2797-809. [PMID: 24877630 PMCID: PMC4098003 DOI: 10.1210/en.2014-1046] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PTH upregulates the expression of the receptor activator of nuclear factor κB ligand (Rankl) in cells of the osteoblastic lineage, but the precise differentiation stage of the PTH target cell responsible for RANKL-mediated stimulation of bone resorption remains undefined. We report that constitutive activation of PTH receptor signaling only in osteocytes in transgenic mice (DMP1-caPTHR1) was sufficient to increase Rankl expression and bone resorption. Resorption in DMP1-caPTHR1 mice crossed with mice lacking the distal control region regulated by PTH in the Rankl gene (DCR(-/-)) was similar to DMP1-caPTHR1 mice at 1 month of age, but progressively declined to reach values undistinguishable from wild-type (WT) mice at 5 months of age. Moreover, DMP1-caPTHR1 mice exhibited low tissue material density and increased serum alkaline phosphatase activity at 5 month of age, and these indices of high remodeling were partially and totally corrected in compound DMP1-caPTHR1;DCR(-/-) male mice, and less affected in female mice. Rankl expression in bones from DMP1-caPTHR1 mice was elevated at both 1 and 5 months of age, whereas it was high, similar to DMP1-caPTHR1 mice at 1 month, but low, similar to WT levels at 5 months in compound mice. Moreover, PTH increased Rankl and decreased Sost and Opg expression in ex vivo bone organ cultures established from WT mice, but only regulated Sost and Opg expression in cultures from DCR(-/-) mice. PTH also increased RANKL expression in osteocyte-containing primary cultures of calvarial cells, in isolated murine osteocytes, and in WT but not in DCR(-/-) osteocyte-enriched bones. Thus, PTH upregulates Rankl expression in osteocytes in vitro, ex vivo and in vivo, and resorption induced by PTH receptor signaling in the adult skeleton requires direct regulation of the Rankl gene in osteocytes.
Collapse
Affiliation(s)
- Abdullah N Ben-awadh
- Departments of Anatomy and Cell Biology (A.N.B., J.D.-C., X.T., K.K., M.R.A., L.I.P., T.B.) and Medicine (T.B.), Division of Endocrinology, Indiana University School of Medicine, and Roudebush Veterans Administration Medical Center (J.D.-C., L.I.P., T.B.), Indianapolis, Indiana 46202
| | | | | | | | | | | | | |
Collapse
|
32
|
Charles JF, Aliprantis AO. Osteoclasts: more than 'bone eaters'. Trends Mol Med 2014; 20:449-59. [PMID: 25008556 PMCID: PMC4119859 DOI: 10.1016/j.molmed.2014.06.001] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/28/2014] [Accepted: 06/02/2014] [Indexed: 02/08/2023]
Abstract
As the only cells definitively shown to degrade bone, osteoclasts are key mediators of skeletal diseases including osteoporosis. Bone-forming osteoblasts, and hematopoietic and immune system cells, each influence osteoclast formation and function, but the reciprocal impact of osteoclasts on these cells is less well appreciated. We highlight here the functions that osteoclasts perform beyond bone resorption. First, we consider how osteoclast signals may contribute to bone formation by osteoblasts and to the pathology of bone lesions such as fibrous dysplasia and giant cell tumors. Second, we review the interaction of osteoclasts with the hematopoietic system, including the stem cell niche and adaptive immune cells. Connections between osteoclasts and other cells in the bone microenvironment are discussed within a clinically relevant framework.
Collapse
Affiliation(s)
- Julia F Charles
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Antonios O Aliprantis
- Department of Medicine, Division of Rheumatology, Allergy, and Immunology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
33
|
Miller PD. Bone Disease in CKD: A Focus on Osteoporosis Diagnosis and Management. Am J Kidney Dis 2014; 64:290-304. [DOI: 10.1053/j.ajkd.2013.12.018] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 12/27/2013] [Indexed: 11/11/2022]
|
34
|
Bialek P, Parkington J, Li X, Gavin D, Wallace C, Zhang J, Root A, Yan G, Warner L, Seeherman HJ, Yaworsky PJ. A myostatin and activin decoy receptor enhances bone formation in mice. Bone 2014; 60:162-71. [PMID: 24333131 DOI: 10.1016/j.bone.2013.12.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 11/25/2013] [Accepted: 12/03/2013] [Indexed: 01/11/2023]
Abstract
Myostatin is a member of the bone morphogenetic protein/transforming growth factor-β (BMP/TGFβ) super-family of secreted differentiation factors. Myostatin is a negative regulator of muscle mass as shown by increased muscle mass in myostatin deficient mice. Interestingly, these mice also exhibit increased bone mass suggesting that myostatin may also play a role in regulating bone mass. To investigate the role of myostatin in bone, young adult mice were administered with either a myostatin neutralizing antibody (Mstn-mAb), a soluble myostatin decoy receptor (ActRIIB-Fc) or vehicle. While both myostatin inhibitors increased muscle mass, only ActRIIB-Fc increased bone mass. Bone volume fraction (BV/TV), as determined by microCT, was increased by 132% and 27% in the distal femur and lumbar vertebrae, respectively. Histological evaluation demonstrated that increased BV/TV in both locations was attributed to increased trabecular thickness, trabecular number and bone formation rate. Increased BV/TV resulted in enhanced vertebral maximum compressive force compared to untreated animals. The fact that ActRIIB-Fc, but not Mstn-mAb, increased bone volume suggested that this soluble decoy receptor may be binding a ligand other than myostatin, that plays a role in regulating bone mass. This was confirmed by the significant increase in BV/TV in myostatin deficient mice treated with ActRIIB-Fc. Of the other known ActRIIB-Fc ligands, BMP3 has been identified as a negative regulator of bone mass. However, BMP3 deficient mice treated with ActRIIB-Fc showed similar increases in BV/TV as wild type (WT) littermates treated with ActRIIB-Fc. This result suggests that BMP3 neutralization is not the mechanism responsible for increased bone mass. The results of this study demonstrate that ActRIIB-Fc increases both muscle and bone mass in mice. Therefore, a therapeutic that has this dual activity represents a potential approach for the treatment of frailty.
Collapse
Affiliation(s)
- P Bialek
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA.
| | - J Parkington
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA
| | - X Li
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA
| | - D Gavin
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA
| | - C Wallace
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA
| | - J Zhang
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA
| | - A Root
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA
| | - G Yan
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA
| | - L Warner
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA
| | - H J Seeherman
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA
| | - P J Yaworsky
- Biotherapeutics Research and Development, Pfizer Inc., 200 CambridgePark Drive, Cambridge, MA 02140, USA
| |
Collapse
|