1
|
Szudy-Szczyrek A, Mlak R, Pigoń-Zając D, Krupski W, Mazurek M, Tomczak A, Chromik K, Górska A, Koźlik P, Juda A, Kokoć A, Dubaj M, Sacha T, Niedoszytko M, Helbig G, Szczyrek M, Szumiło J, Małecka-Massalska T, Hus M. Role of sclerostin in mastocytosis bone disease. Sci Rep 2025; 15:161. [PMID: 39747949 PMCID: PMC11697018 DOI: 10.1038/s41598-024-83851-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Mastocytosis is a heterogeneous group of disorders, characterized by accumulation of clonal mast cells which can infiltrate several organs, most often spine (70%). The pathogenesis of mastocytosis bone disease is poorly understood. The main aim of the study was to investigate whether neoplastic mast cells may be the source of sclerostin and whether there is an association between sclerostin and selected bone remodeling markers with mastocytosis related bone disease. We assessed sclerostin, bioactive sclerostin, and SOST gene expression in HMC-1.2 human mast cell culture supernatants and plasma of SM patients (n = 39). We showed that human mast cells can secrete sclerostin, and after their stimulation with IL-6, there is a significant increase in SOST gene expression. We observed significantly higher levels of sclerostin in patients diagnosed with more advanced disease. We observed a statistically significant correlation between concentations of sclerostin and its bioactive form and the concentration of alkaline phosphatase (ALP), and between sclerostin and interleukin-6 (IL-6). We observed that significantly higher sclerostin concentrations are present in patients with increased sclerosis of the spongy bone. Sclerostin may serve as a marker of more advanced disease and bone disease in mastocytosis. Further studies are justified to evaluate its role in mastocytosis.
Collapse
Affiliation(s)
- Aneta Szudy-Szczyrek
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Staszica Street 11, 20-081, Poland
| | - Radosław Mlak
- Department of Laboratory Diagnostics, Medical University of Lublin, Lublin, Poland
| | | | - Witold Krupski
- Department of Medical Radiology, Medical University of Lublin, Lublin, Poland
| | - Marcin Mazurek
- Department of Human Physiology, Medical University of Lublin, Lublin, Poland
| | - Aleksandra Tomczak
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Staszica Street 11, 20-081, Poland
| | - Karolina Chromik
- Department of Hematology and Bone Marrow Transplantation, Medical University of Silesia in Katowice, Katowice, Poland
| | - Aleksandra Górska
- Department of Pulmonology and Allergology, Medical University of Gdańsk, Gdańsk, Poland
| | - Paweł Koźlik
- Chair of Hematology, Jagiellonian University, Kraków, Poland
| | - Adrian Juda
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Staszica Street 11, 20-081, Poland
| | - Anna Kokoć
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Staszica Street 11, 20-081, Poland
| | - Maciej Dubaj
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Staszica Street 11, 20-081, Poland.
| | - Tomasz Sacha
- Chair of Hematology, Jagiellonian University, Kraków, Poland
| | - Marek Niedoszytko
- Department of Pulmonology and Allergology, Medical University of Gdańsk, Gdańsk, Poland
| | - Grzegorz Helbig
- Department of Hematology and Bone Marrow Transplantation, Medical University of Silesia in Katowice, Katowice, Poland
| | - Michał Szczyrek
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Justyna Szumiło
- Department of Clinical Pathomorphology, Medical University of Lublin, Lublin, Poland
| | | | - Marek Hus
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Staszica Street 11, 20-081, Poland
| |
Collapse
|
2
|
Anastasilakis AD, Yavropoulou MP, Palermo A, Makras P, Paccou J, Tabacco G, Naciu AM, Tsourdi E. Romosozumab versus parathyroid hormone receptor agonists: which osteoanabolic to choose and when? Eur J Endocrinol 2024; 191:R9-R21. [PMID: 38938063 DOI: 10.1093/ejendo/lvae076] [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: 04/26/2024] [Revised: 05/31/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Osteoanabolic agents are used as a first line treatment in patients at high fracture risk. The PTH receptor 1 (PTH1R) agonists teriparatide (TPTD) and abaloparatide (ABL) increase bone formation, bone mineral density (BMD), and bone strength by activating PTH receptors on osteoblasts. Romosozumab (ROMO), a humanized monoclonal antibody against sclerostin, dramatically but transiently stimulates bone formation and persistently reduces bone resorption. Osteoanabolic agents increase BMD and bone strength while being more effective than antiresorptives in reducing fracture risk in postmenopausal women. However, direct comparisons of the antifracture benefits of osteoanabolic therapies are limited. In a direct comparison of TPTD and ABL, the latter resulted in greater BMD increases at the hip. While no differences in vertebral or non-vertebral fracture risk were observed between the two drugs, ABL led to a greater reduction of major osteoporotic fractures. Adverse event profiles were similar between the two agents except for hypercalcemia, which occurred more often with TPTD. No direct comparisons of fracture risk reduction between ROMO and the PTH1R agonists exist. Individual studies have shown greater increases in BMD and bone strength with ROMO compared with TPTD in treatment-naive women and in women previously treated with bisphosphonates. Some safety aspects, such as a history of tumor precluding the use of PTH1R agonists, and a history of major cardiovascular events precluding the use of ROMO, should also be considered when choosing between these agents. Finally, convenience of administration, reimbursement by national health systems and length of clinical experience may influence patient choice.
Collapse
Affiliation(s)
| | - Maria P Yavropoulou
- Endocrinology Unit, 1st Department of Propaedeutic and Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Laikon University Hospital of Athens, Athens 115 27, Greece
| | - Andrea Palermo
- Unit of Metabolic Bone and Thyroid Diseases, Fondazione Policlinico Universitario Campus Bio-Medico, Rome 00128, Italy
- Unit of Endocrinology and Diabetes, Campus Bio-Medico University of Rome, Rome 00128, Italy
| | - Polyzois Makras
- Department of Medical Research, 251 Hellenic Air Force & VA General Hospital, Athens 115 25, Greece
| | - Julien Paccou
- Department of Rheumatology, CHU Lille, Lille 59000, France
| | - Gaia Tabacco
- Unit of Metabolic Bone and Thyroid Diseases, Fondazione Policlinico Universitario Campus Bio-Medico, Rome 00128, Italy
- Unit of Endocrinology and Diabetes, Campus Bio-Medico University of Rome, Rome 00128, Italy
| | - Anda Mihaela Naciu
- Unit of Metabolic Bone and Thyroid Diseases, Fondazione Policlinico Universitario Campus Bio-Medico, Rome 00128, Italy
- Unit of Endocrinology and Diabetes, Campus Bio-Medico University of Rome, Rome 00128, Italy
| | - Elena Tsourdi
- Department of Medicine III, Technische Universität Dresden, Dresden 01307, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden 01307, Germany
| |
Collapse
|
3
|
Li M, Hasan AA, Chu C, Hocher JG, Liu Y, Zhang X, Chen X, Yard B, Krämer BK, Hocher B. Only bioactive forms of PTH (n-oxPTH and Met18(ox)-PTH) inhibit synthesis of sclerostin - evidence from in vitro and human studies. Pflugers Arch 2024; 476:889-899. [PMID: 38393416 PMCID: PMC11139748 DOI: 10.1007/s00424-024-02928-x] [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: 09/28/2023] [Revised: 01/30/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024]
Abstract
Sclerostin (SOST) is produced by osteocytes and is known as a negative regulator of bone homeostasis. Parathyroid hormone (PTH) regulates calcium, phosphate as well as vitamin D metabolism, and is a strong inhibitor of SOST synthesis in vitro and in vivo. PTH has two methionine amino acids (positions 8 and 18) which can be oxidized. PTH oxidized at Met18 (Met18(ox)-PTH) continues to be bioactive, whereas PTH oxidized at Met8 (Met8(ox)-PTH) or PTH oxidized at Met8 and Met18 (Met8, Met18(di-ox)-PTH) has minor bioactivity. How non-oxidized PTH (n-oxPTH) and oxidized forms of PTH act on sclerostin synthesis is unknown. The effects of n-oxPTH and oxidized forms of PTH on SOST gene expression were evaluated in UMR106 osteoblast-like cells. Moreover, we analyzed the relationship of SOST with n-oxPTH and all forms of oxPTH in 516 stable kidney transplant recipients using an assay system that can distinguish in clinical samples between n-oxPTH and the sum of all oxidized PTH forms (Met8(ox)-PTH, Met18(ox)-PTH, and Met8, Met18(di-ox)-PTH). We found that both n-oxPTH and Met18(ox)-PTH at doses of 1, 3, 20, and 30 nmol/L significantly inhibit SOST gene expression in vitro, whereas Met8(ox)-PTH and Met8, Met18(di-ox)-PTH only have a weak inhibitory effect on SOST gene expression. In the clinical cohort, multivariate linear regression showed that only n-oxPTH, but not intact PTH (iPTH) nor oxPTH, is independently associated with circulating SOST after adjusting for known confounding factors. In conclusion, only bioactive PTH forms such as n-oxPTH and Met18(ox)-PTH, inhibit SOST synthesis.
Collapse
Affiliation(s)
- Mei Li
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Ahmed A Hasan
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Chang Chu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Johann-Georg Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Yvonne Liu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Xiaoli Zhang
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Xin Chen
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Benito Yard
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.
- Reproductive, Genetic Hospital of CITIC-Xiangya, Changsha, China.
- Institute of Medical Diagnostics, IMD Berlin-Potsdam, Berlin, Germany.
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Jain A, Sharma P, N S, Sharma K, Gupta S. Gene Polymorphism Influencing Persistent Bone Resorption in Brown Tumor: A Case Series with Possible Pathogenesis and Potential Therapeutic Approach. Indian J Otolaryngol Head Neck Surg 2024; 76:1178-1182. [PMID: 38440495 PMCID: PMC10908929 DOI: 10.1007/s12070-023-04203-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/27/2023] [Indexed: 03/06/2024] Open
Abstract
Brown tumor represents a terminal stage of bone remodeling process due to an imbalance between osteoclastic and osteoblastic activity. It represents a reparative cellular process, rather than a neoplastic process mostly associated with primary or secondary hyperparathyroidism. Although parathyroidectomy is the first treatment of choice for brown tumors, several cases don't resolve even after normalization of parathyroid hormone levels which leads to surgical intervention. Therefore, to avoid multiple bone surgeries in the same patient, it is crucial to have a conservative approach like targeted therapy which could block certain molecules involved in bone resorption. In this string, we have recognized and quantified three molecules namely sclerostin, MCP-1 and CD73 in brown tumors and correlated their expression with bone resorption pathogenesis and potential therapeutic approach.
Collapse
Affiliation(s)
- Ayushi Jain
- Dept of Oral Pathology, Microbiology and Forensic Odontology, King George’s Medical University, Lucknow, 226003 UP India
| | - Pooja Sharma
- Dept of Oral Pathology, Microbiology and Forensic Odontology, King George’s Medical University, Lucknow, 226003 UP India
| | - Sivakumar N
- Dept of Oral Pathology, Microbiology and Forensic Odontology, All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Kriti Sharma
- Dept of Oral Pathology, Microbiology and Forensic Odontology, King George’s Medical University, Lucknow, 226003 UP India
| | - Shalini Gupta
- Dept of Oral Pathology, Microbiology and Forensic Odontology, King George’s Medical University, Lucknow, 226003 UP India
| |
Collapse
|
5
|
Dincel AS, Jørgensen NR. New Emerging Biomarkers for Bone Disease: Sclerostin and Dickkopf-1 (DKK1). Calcif Tissue Int 2023; 112:243-257. [PMID: 36165920 DOI: 10.1007/s00223-022-01020-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/26/2022] [Indexed: 01/25/2023]
Abstract
A healthy skeleton depends on a continuous renewal and maintenance of the bone tissue. The process of bone remodeling is highly controlled and consists of a fine-tuned balance between bone formation and bone resorption. Biochemical markers of bone turnover are already in use for monitoring diseases and treatment involving the skeletal system, but novel biomarkers reflecting specific biological processes in bone and interacting tissues may prove useful for diagnostic, prognostic, and monitoring purposes. The Wnt-signaling pathway is one of the most important pathways controlling bone metabolism and consequently the action of inhibitors of the pathway such as sclerostin and Dickkopf-related protein 1 (DKK1) have crucial roles in controlling bone formation and resorption. Thus, they might be potential markers for clinical use as they reflect a number of physiological and pathophysiological events in bone and in the cross-talk with other tissues in the human body. This review focuses on the clinical utility of measurements of circulating sclerostin and DKK1 levels based on preanalytical and analytical considerations and on evidence obtained from published clinical studies. While accumulating evidence points to clear associations with a number of disease states for the two markers, and thus, the potential for especially sclerostin as a biochemical marker that may be used clinically, the lack of standardization or harmonization of the assays still hampers the clinical utility of the markers.
Collapse
Affiliation(s)
- Aylin Sepinci Dincel
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, Ankara, Turkey
- Department of Clinical Biochemistry, Rigshospitalet, Valdemar Hansens Vej 13 Glostrup, 2600, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niklas Rye Jørgensen
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, Ankara, Turkey.
- Department of Clinical Biochemistry, Rigshospitalet, Valdemar Hansens Vej 13 Glostrup, 2600, Copenhagen, Denmark.
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
6
|
Burr AM, Zuckerman PC, Castillo AB, Partridge NC, Parekkadan B. Bioactive, full-length parathyroid hormone delivered using an adeno-associated viral vector. Exp Biol Med (Maywood) 2022; 247:1885-1897. [PMID: 35666091 PMCID: PMC9742744 DOI: 10.1177/15353702221097087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Delivering the parathyroid hormone (PTH) gene has been attempted preclinically in a handful of studies, but delivering full-length PTH (1-84) using adeno-associated viral (AAV) vectors has not. Given the difficulty in achieving therapeutic levels of secreted proteins using gene therapy, this study seeks to determine the feasibility of doing so with PTH. An AAV vector was used to deliver human PTH driven by a strong promoter. We demonstrate the ability to secrete full-length PTH from various cell types in vitro. PTH secretion from hepatocytes was measured over time and a fluorescent marker was used to compare the secretion rate of PTH in various cell types. Potency was measured by the ability of PTH to act on the PTH receptors of osteosarcoma cells and induced proliferation. PTH showed potency in vitro by inducing proliferation in two osteosarcoma cell lines. In vivo, AAV was administered systemically in immunocompromised mice which received xenografts of osteosarcoma cells. Animals that received the highest dose of AAV-PTH had higher liver and plasma concentrations of PTH. All dosing groups achieved measurable plasma concentrations of human PTH that were above the normal range. The high-dose group also had significantly larger tumors compared to control groups on the final day of the study. The tumors also showed dose-dependent differences in morphology. When looking at endocrine signaling and endogenous bone turnover, we observed a significant difference in tibial growth plate width in animals that received the high-dose AAV as well as dose-dependent changes in blood biomarkers related to PTH. This proof-of-concept study shows promise for further exploration of an AAV gene therapy to deliver full-length PTH for hypoparathyroidism. Additional investigation will determine efficacy in a disease model, but data shown establish bioactivity in well-established models of osteosarcoma.
Collapse
Affiliation(s)
- Alexandra M Burr
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Pamela Cabahug Zuckerman
- Department of Orthopedic Surgery, NYU Langone Health, New York University, New York, NY 10016, USA.,Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 11201, USA.,Rehabilitation Research and Development, Veterans Affairs New York Harbor Healthcare System, New York, NY 11209, USA
| | - Alesha B Castillo
- Department of Orthopedic Surgery, NYU Langone Health, New York University, New York, NY 10016, USA.,Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 11201, USA.,Rehabilitation Research and Development, Veterans Affairs New York Harbor Healthcare System, New York, NY 11209, USA
| | - Nicola C Partridge
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
7
|
Nagata Y, Imanishi Y, Tateishi T, Miyaoka D, Kurajoh M, Arnold A, Emoto M. Parathyroid Hormone Regulates Circulating Levels of Sclerostin and FGF23 in a Primary Hyperparathyroidism Model. J Endocr Soc 2022; 6:bvac027. [PMID: 35284773 PMCID: PMC8907412 DOI: 10.1210/jendso/bvac027] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Indexed: 11/21/2022] Open
Abstract
Parathyroid hormone (PTH) increases fibroblast growth factor 23 (FGF23), mediated both by protein kinase A (PKA) and Wnt signaling, and decreases expression of sclerostin, a Wnt antagonist derived from osteocytes. Patients with primary hyperparathyroidism (PHPT) have lower serum sclerostin levels than healthy controls, consistent with the idea of SOST downregulation by PTH. Nevertheless, the relationship between FGF23 and sclerostin in PHPT is still unclear. We examined this issue in a mouse model of PHPT. PHPT mice had increased FGF23 and decreased sclerostin expression in calvaria and in their serum concentrations compared with wild-type (WT) mice. In UMR106 osteoblasts, PTH increased Fgf23 expression and decreased Sost expression, as well as forskolin, a PKA agonist, whereas inhibition of PKA reversed the changes in Fgf23 and Sost expression, stimulated by PTH. Sclerostin treatment had no effect on Fgf23 expression, but when it was added together with PTH, it statistically significantly abrogated the increase in Fgf23 expression. By contrast, there was no statistically significant correlation between serum FGF23 and sclerostin, whereas PTH was positively and negatively correlated with serum FGF23 and sclerostin, respectively. These results indicate that the high level of PTH in PHPT mice leads to increased FGF23 and decreased sclerostin expression in serum and calvaria. A decrease of sclerostin may further augment FGF23 in vitro; however, there was no statistically significant association between circulating FGF23 and sclerostin. It is suggested that the pathogenesis of increased FGF23 expression in PHPT mice may be modified by not only sclerostin, but also other regulatory factors modulated by PTH.
Collapse
Affiliation(s)
- Yuki Nagata
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
- Department of Vascular Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Vascular Science Center for Translational Research, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Yasuo Imanishi
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tomomi Tateishi
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Daichi Miyaoka
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masafumi Kurajoh
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Andrew Arnold
- Division of Endocrinology and Metabolism, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Masanori Emoto
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
8
|
Claessen KMJA, Pelsma ICM, Kroon HM, van Lierop AH, Pereira AM, Biermasz NR, Appelman-Dijkstra NM. Low sclerostin levels after long-term remission of acromegaly. Endocrine 2022; 75:228-238. [PMID: 34448099 PMCID: PMC8763730 DOI: 10.1007/s12020-021-02850-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/11/2021] [Indexed: 10/30/2022]
Abstract
PURPOSE Bone health is compromised in acromegaly resulting in vertebral fractures (VFs), regardless of biochemical remission. Sclerostin is a negative inhibitor of bone formation and is associated with increased fracture risk in the general population. Therefore, we compared sclerostin concentrations between well-controlled acromegaly patients and healthy controls, and assessed its relationship with bone mineral density (BMD), and VFs in acromegaly. METHODS Seventy-nine patients (mean age 58.9 ± 11.4 years, 49% women) with controlled acromegaly, and 91 healthy controls (mean age 51.1 ± 16.9 years, 59% women) were included. Plasma sclerostin levels (pg/mL) in patients were measured with an ELISA assay, whereas in controls, serum levels were converted to plasma levels by multiplication with 3.6. In patients, VFs were radiographically assessed, and BMD was assessed using dual X-ray absorptiometry. RESULTS Median sclerostin concentration in controlled acromegaly patients was significantly lower than in healthy controls (104.5 pg/mL (range 45.7-234.7 pg/mL) vs 140.0 pg/mL (range 44.8-401.6 pg/mL), p < 0.001). Plasma sclerostin levels were not related to age, current growth hormone (GH) or insulin-like factor-1 (IGF-1) levels, gonadal state, treatment modality, remission duration, or BMD, VF presence, severity or progression. CONCLUSION Patients with long-term controlled acromegaly have lower plasma sclerostin levels than healthy controls, as a reflection of decreased osteocyte activity. Further longitudinal studies are needed to establish the course of sclerostin during different phases of disease and its exact effects in acromegalic osteopathy.
Collapse
Affiliation(s)
- Kim M J A Claessen
- Department of Medicine, Division of Endocrinology, and Center for Endocrine Tumors Leiden, Leiden, The Netherlands.
- Department of Medicine, Division of Endocrinology, and Center for Bone Quality, Leiden, The Netherlands.
| | - Iris C M Pelsma
- Department of Medicine, Division of Endocrinology, and Center for Endocrine Tumors Leiden, Leiden, The Netherlands
| | - Herman M Kroon
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Antoon H van Lierop
- Department of Medicine, Division of Endocrinology, and Center for Bone Quality, Leiden, The Netherlands
| | - Alberto M Pereira
- Department of Medicine, Division of Endocrinology, and Center for Endocrine Tumors Leiden, Leiden, The Netherlands
| | - Nienke R Biermasz
- Department of Medicine, Division of Endocrinology, and Center for Endocrine Tumors Leiden, Leiden, The Netherlands
| | - Natasha M Appelman-Dijkstra
- Department of Medicine, Division of Endocrinology, and Center for Endocrine Tumors Leiden, Leiden, The Netherlands
- Department of Medicine, Division of Endocrinology, and Center for Bone Quality, Leiden, The Netherlands
| |
Collapse
|
9
|
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
|
10
|
Assessment of Clinical Utility of Assaying FGF-23, Klotho Protein, Osteocalcin, NTX, and Sclerostin in Patients with Primary Hyperparathyroidism. J Clin Med 2021; 10:jcm10143089. [PMID: 34300255 PMCID: PMC8303928 DOI: 10.3390/jcm10143089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/09/2021] [Accepted: 07/11/2021] [Indexed: 12/28/2022] Open
Abstract
The purpose of this study was to assess the clinical usefulness of assaying the fibroblast growth factor (FGF-23), Klotho, osteocalcin, N-terminal telopeptide of type I collagen (NTX), and sclerostin levels in patients with primary hyperparathyroidism (PHPT) as markers of bone damage as well as for surgical treatment success. Seventeen patients with hypercalcemic PHPT and normal kidney function were studied. In all patients, PTH (parathormone), serum calcium, and creatinine were performed before and six months after parathyroidectomy (PTX). The studied group included patients whose PTH and calcium concentrations normalized post-operatively and with confirmed histopathological diagnosis. The control group consisted of nine age-matched healthy volunteers. The PHPT patients had elevated concentrations of FGF-23, osteocalcin, and NTX and reduced levels of sclerostin, as compared to the control group. After PTX, osteocalcin, NTX, and sclerostin levels normalized. The plasma values of FGF-23 decreased significantly, but remained higher than in healthy subjects. Serum Klotho protein levels did not differ significantly in the two groups. These results suggest that osteocalcin and NTX may potentially be considered as markers of PHPT progression. Additionally, serum normalization of osteocalcin, NTX, and sclerostin might be considered as indicators of PTX success. On the other hand, FGF-23 can represent a parameter reflecting the degree of calcium–phosphate imbalance in PHPT patients, but its usefulness in monitoring the effects of PTX requires further research. The clinical utility of assaying Klotho in PHPT remains to be confirmed.
Collapse
|
11
|
Abstract
Skeletal integrity is maintained by a meticulous balance between bone resorption and bone formation, and recent studies have revealed the essential role of canonical Wnt signaling pathways in maintaining skeletal homeostasis. The SOST gene, which encodes sclerostin, a member of Dan family glycoproteins, was originally identified as the gene responsible for two sclerosing bone dysplasias, sclerosteosis and van Buchem disease. Sclerostin is highly expressed by osteocytes, negatively regulates canonical Wnt signaling pathways by binding to low-density lipoprotein receptor-related protein (LRP) 5/6, and suppresses osteoblast differentiation and/or function. Romosozumab, a specific anti-sclerostin antibody, inhibits sclerostin-LRP5/6 interactions and indirectly activates canonical Wnt signaling pathways and bone formation. This review focuses on the mechanism of action of sclerostin and summarizes clinical studies that demonstrated the efficacy of romosozumab to increase bone mineral density and reduce osteoporotic fractures, as well as its cardiovascular safety.
Collapse
Affiliation(s)
- Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
12
|
Yang YB, Goshtasebi A, van Lierop AH, Kalidasan D, Hitchcock CL, Prior JC. Effects of progesterone therapy on serum sclerostin levels in healthy menopausal women: a 3-month randomized, placebo-controlled clinical trial. Osteoporos Int 2020; 31:2243-2250. [PMID: 32564093 DOI: 10.1007/s00198-020-05505-x] [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: 12/17/2019] [Accepted: 06/15/2020] [Indexed: 10/24/2022]
Abstract
UNLABELLED Sclerostin, a natural hormone made in bone, suppresses bone formation. Sclerostin is also decreased by estrogen. Progesterone, estrogen's menstrual partner, stimulates bone formation. It is unclear whether progesterone influences sclerostin. This study showed that progesterone did not change sclerostin using serum remaining from a randomized progesterone hot flush therapy trial. INTRODUCTION Progesterone and sclerostin are both endogenous hormones acting through osteoblast-origin cells and promote or suppress bone formation, respectively. Estradiol suppresses sclerostin, but progesterone, its menstrual cycle partner hormone, has unclear sclerostin relationships. We postulated that progesterone therapy would influence serum sclerostin levels. METHODS We obtained sclerostin levels for an ethics-approved post hoc analysis. Fasting sclerostin was measured in all remaining sera from a previous 12-week randomized controlled trial (RCT) of oral micronized progesterone (progesterone) for menopausal (> 1 year after last flow) vasomotor symptoms (VMS). Women in the RCT took 300 mg progesterone at bedtime or placebo (1:1) in a trial showing progesterone significantly decreased VMS. RESULTS Participants were healthy menopausal, primarily Caucasian (91.2%) community-dwelling women (± SD), 55.2 ± 4.6 years old with BMI 24.9 ± 2.9 kg/m2. The baseline sclerostin level in 60 women was 28.41 ± 10.47 pmol/L. Baseline sclerostin was not correlated with the run-in VMS score (r = 0.143, P = 0.294). Paired baseline and 12-week RCT data for 52 women showed serum sclerostin levels did not change related to experimental therapy (P = 0.504). Changes in final sclerostin values adjusted for baseline were progesterone (- 1.07 ± 7.96 pmol/L) and placebo (- 2.64 ± 8.70 pmol/L). In observational data (n = 60), baseline sclerostin levels correlated with the General Framingham Cardiovascular (CVD) Risk score (r = - 0.398, P = 0.003) and self-reported health by SF-36 quality of life instrument (QoL, r = - 0.331, P = 0.016). CONCLUSION Physiological oral micronized progesterone did not stimulate nor suppress serum sclerostin levels based on post hoc analysis of RCT data. Exploratory results, however, showed sclerostin negatively correlated with CVD risk and QoL. ClinicalTrials.gov #NCT0146469.
Collapse
Affiliation(s)
- Y B Yang
- Department of Medicine, Division of Endocrinology, Centre for Menstrual Cycle and Ovulation Research (CeMCOR), University of British Columbia, Vancouver, British Columbia, Canada
| | - A Goshtasebi
- Department of Medicine, Division of Endocrinology, Centre for Menstrual Cycle and Ovulation Research (CeMCOR), University of British Columbia, Vancouver, British Columbia, Canada
| | - A H van Lierop
- Center for Bone Quality, Leiden University Medical Center, Leiden, The Netherlands
| | - D Kalidasan
- Department of Medicine, Division of Endocrinology, Centre for Menstrual Cycle and Ovulation Research (CeMCOR), University of British Columbia, Vancouver, British Columbia, Canada
| | - C L Hitchcock
- Department of Medicine, Division of Endocrinology, Centre for Menstrual Cycle and Ovulation Research (CeMCOR), University of British Columbia, Vancouver, British Columbia, Canada
| | - J C Prior
- Department of Medicine, Division of Endocrinology, Centre for Menstrual Cycle and Ovulation Research (CeMCOR), University of British Columbia, Vancouver, British Columbia, Canada.
- School of Population and Public Health, University of British Columbia, Vancouver, Canada.
- British Columbia Women's Health Research Institute, Vancouver, Canada.
- Department of Medicine, Division of Endocrinology and Metabolism, University of British Columbia, 2775 Laurel Street, Suite 4111, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
13
|
Osteocyte-Related Cytokines Regulate Osteoclast Formation and Bone Resorption. Int J Mol Sci 2020; 21:ijms21145169. [PMID: 32708317 PMCID: PMC7404053 DOI: 10.3390/ijms21145169] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/16/2020] [Accepted: 07/20/2020] [Indexed: 01/18/2023] Open
Abstract
The process of bone remodeling is the result of the regulated balance between bone cell populations, namely bone-forming osteoblasts, bone-resorbing osteoclasts, and the osteocyte, the mechanosensory cell type. Osteoclasts derived from the hematopoietic stem cell lineage are the principal cells involved in bone resorption. In osteolytic diseases such as rheumatoid arthritis, periodontitis, and osteoporosis, the balance is lost and changes in favor of bone resorption. Therefore, it is vital to elucidate the mechanisms of osteoclast formation and bone resorption. It has been reported that osteocytes express Receptor activator of nuclear factor κΒ ligand (RANKL), an essential factor for osteoclast formation. RANKL secreted by osteocytes is the most important factor for physiologically supported osteoclast formation in the developing skeleton and in pathological bone resorption such as experimental periodontal bone loss. TNF-α directly enhances RANKL expression in osteocytes and promotes osteoclast formation. Moreover, TNF-α enhances sclerostin expression in osteocytes, which also increases osteoclast formation. These findings suggest that osteocyte-related cytokines act directly to enhance osteoclast formation and bone resorption. In this review, we outline the most recent knowledge concerning bone resorption-related cytokines and discuss the osteocyte as the master regulator of bone resorption and effector in osteoclast formation.
Collapse
|
14
|
Effect of fluoride on osteocyte-driven osteoclastic differentiation. Toxicology 2020; 436:152429. [PMID: 32156525 DOI: 10.1016/j.tox.2020.152429] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 11/24/2022]
Abstract
Excessive systemic uptake of inorganic fluorides causes disturbances of bone homeostasis. The mechanism of skeletal fluorosis is still uncertain. This study aimed to study the effect of fluoride on osteocyte-driven osteoclastogenesis and probe into the role of PTH in this process. IDG-SW3 cells seeded in collagen-coated constructs were developed into osteocyte-like cells through induction of mineral agents. Then, osteocyte-like cells were exposed to fluoride in the presence or absence of parathyroid hormone (PTH). Cell viability and their capacity to produce receptor activator of nuclear factor kappa-B ligand (RANKL), osteoprotegerin (OPG) and sclerostin (SOST) were detected by MTT and Western blot assays, respectively. Finally, a transwell coculture system using osteocyte-like cells seeded in the low compartment, and osteoclast precursors added in the inserts was developed to observe the osteocyte-driven osteoclasogenesis response to fluoride with or without PTH, and the expression of molecules involved in this mechanism were measure by real time RT-PCR. Results showed that osteocytes withstood a toxic dose of fluoride, and yet PTH administration significantly reduced osteocytes viability. PTH amplified the effect of fluoride on the expression of osteoclastogenesis-related molecules in osteocyte, but did not enlarged the stimulating effect of fluoride on osteoclastogenesis drove by osteocyte coculture. Gene expression levels of TRAP, RANK, JNK and NFAtc1 significantly increased in fluoride affected osteoclast precursor cocultured with osteocyte-like cells. The impact of fluoride on osteocyte-driven osteoclast differentiation was stronger than that of PTH. In conclusion, osteocyte played a pivotal role on the mechanism underlying fluoride-affected osteoclastogenesis in which RANK-JNK-NFATc1 signaling pathway was involved, and PTH had a significant impact in this process.
Collapse
|
15
|
Bouquegneau A, Evenepoel P, Paquot F, Malaise O, Cavalier E, Delanaye P. Sclerostin within the chronic kidney disease spectrum. Clin Chim Acta 2019; 502:84-90. [PMID: 31866333 DOI: 10.1016/j.cca.2019.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 01/08/2023]
Abstract
Sclerostin is sometimes presented as a promising biomarker in assessing bone health both in the general population and chronic kidney disease patients. However, it is still unclear whether it has any true added value compared to existing bone biomarkers in predicting bone turnover and/or bone density in chronic kidney disease patients. A wealth of papers has been published to evaluate the association between sclerostin and vascular calcifications development or even as prognostic biomarker for mortality, but often with conflicting results. Standardization and harmonization of analytical techniques is a prerequisite to advance clinical knowledge in sclerostin.
Collapse
Affiliation(s)
- Antoine Bouquegneau
- Department of Nephrology-Dialysis-Transplantation, University Hospital of Liege (ULg CHU), Liege, Belgium.
| | - Peter Evenepoel
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - François Paquot
- Department of Nephrology-Dialysis-Transplantation, University Hospital of Liege (ULg CHU), Liege, Belgium
| | - Olivier Malaise
- Department of Rheumatology, University Hospital of Liege (ULg CHU), Liege, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, University Hospital of Liege (ULg CHU), Liege, Belgium
| | - Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University Hospital of Liege (ULg CHU), Liege, Belgium
| |
Collapse
|
16
|
De Maré A, Verhulst A, Cavalier E, Delanaye P, Behets GJ, Meijers B, Kuypers D, D’Haese PC, Evenepoel P. Clinical Inference of Serum and Bone Sclerostin Levels in Patients with End-Stage Kidney Disease. J Clin Med 2019; 8:jcm8122027. [PMID: 31756992 PMCID: PMC6947521 DOI: 10.3390/jcm8122027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/10/2019] [Accepted: 11/18/2019] [Indexed: 01/06/2023] Open
Abstract
Mounting evidence indicates that sclerostin, a well-known inhibitor of bone formation, may qualify as a clinically relevant biomarker of chronic kidney disease-related mineral and bone disorder (CKD-MBD), including abnormal mineral and bone metabolism and extraskeletal calcification. For this purpose, in this study we investigate the extent to which circulating sclerostin, skeletal sclerostin expression, bone histomorphometric parameters, and serum markers of bone metabolism associate with each other. Bone biopsies and serum samples were collected in a cohort of 68 end-stage kidney disease (ESKD) patients. Serum sclerostin levels were measured using 4 different commercially available assays. Skeletal sclerostin expression was evaluated on immunohistochemically stained bone sections. Quantitative bone histomorphometry was performed on Goldner stained tissue sections. Different serum markers of bone metabolism were analyzed using in-house techniques or commercially available assays. Despite large inter-assay differences for circulating sclerostin, results obtained with the 4 assays under study closely correlated with each other, whilst moderate significant correlations with skeletal sclerostin expression were also found. Both skeletal and circulating sclerostin negatively correlated with histomorphometric bone and serum parameters reflecting bone formation and turnover. In this study, the unique combined evaluation of bone sclerostin expression, bone histomorphometry, bone biomarkers, and serum sclerostin levels, as assessed by 4 different assays, demonstrated that sclerostin may qualify as a clinically relevant marker of disturbed bone metabolism in ESKD patients.
Collapse
Affiliation(s)
- Annelies De Maré
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; (A.D.M.); (A.V.)
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; (A.D.M.); (A.V.)
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, Domaine du Sart Tilman, 4000 Liège, Belgium;
| | - Pierre Delanaye
- Department of Nephrology, Dialysis, Hypertension, Transplantation, University of Liège, Domaine du Sart Tilman, 4000 Liège, Belgium;
| | - Geert J. Behets
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; (A.D.M.); (A.V.)
| | - Bjorn Meijers
- Nephrology, Division of Internal Medicine, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium; (B.M.); (D.K.); (P.E.)
- Laboratory of Nephrology, Department of Immunology and Microbiology, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
| | - Dirk Kuypers
- Nephrology, Division of Internal Medicine, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium; (B.M.); (D.K.); (P.E.)
- Laboratory of Nephrology, Department of Immunology and Microbiology, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
| | - Patrick C. D’Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; (A.D.M.); (A.V.)
- Correspondence: ; Tel.: +32-326-525-99; Fax: +32-326-525-92
| | - Pieter Evenepoel
- Nephrology, Division of Internal Medicine, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium; (B.M.); (D.K.); (P.E.)
- Laboratory of Nephrology, Department of Immunology and Microbiology, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
| |
Collapse
|
17
|
Kuo TH, Lin WH, Chao JY, Wu AB, Tseng CC, Chang YT, Liou HH, Wang MC. Serum sclerostin levels are positively related to bone mineral density in peritoneal dialysis patients: a cross-sectional study. BMC Nephrol 2019; 20:266. [PMID: 31315601 PMCID: PMC6637583 DOI: 10.1186/s12882-019-1452-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/03/2019] [Indexed: 02/07/2023] Open
Abstract
Background Sclerostin, an antagonist of the Wingless-type mouse mammary tumor virus integration site (Wnt) pathway that regulates bone metabolism, is a potential contributor of chronic kidney disease (CKD)–mineral and bone disorder (MBD), which has various forms of presentation, from osteoporosis to vascular calcification. The positive association of sclerostin with bone mineral density (BMD) has been demonstrated in CKD and hemodialysis (HD) patients but not in peritoneal dialysis (PD) patients. This study assessed the association between sclerostin and BMD in PD patients. Methods Eighty-nine PD patients were enrolled; their sera were collected for measurement of sclerostin and other CKD–MBD-related markers. BMD was also assessed simultaneously. We examined the relationship between sclerostin and each parameter through Spearman correlation analysis and by comparing group data between patients with above- and below-median sclerostin levels. Univariate and multiple logistic regression models were employed to define the most predictive of sclerostin levels in the above-median category. Results Bivariate analysis revealed that sclerostin was correlated with spine BMD (r = 0.271, P = 0.011), spine BMD T-score (r = 0.274, P = 0.010), spine BMD Z-score (r = 0.237, P = 0.027), and intact parathyroid hormone (PTH; r = − 0.357, P < 0.001) after adjustments for age and sex. High BMD, old age, male sex, increased weight and height, diabetes, and high osteocalcin and uric acid levels were observed in patients with high serum sclerostin levels and an inverse relation was noticed between PTH and sclerostin. Univariate logistic regression analysis demonstrated that BMD is positively correlated with above-median sclerostin levels (odds ratio [OR] = 65.61, P = 0.002); the correlation was retained even after multivariate adjustment (OR = 121.5, P = 0.007). Conclusions For the first time, this study demonstrated a positive association between serum sclerostin levels and BMD in the PD population.
Collapse
Affiliation(s)
- Te-Hui Kuo
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Shengli Rd., North Dist, Tainan, 704, Taiwan.,Department and Graduate Institute of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Hung Lin
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jo-Yen Chao
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Shengli Rd., North Dist, Tainan, 704, Taiwan.,Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - An-Bang Wu
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Shengli Rd., North Dist, Tainan, 704, Taiwan.,Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chin-Chung Tseng
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Shengli Rd., North Dist, Tainan, 704, Taiwan
| | - Yu-Tzu Chang
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Shengli Rd., North Dist, Tainan, 704, Taiwan
| | - Hung-Hsiang Liou
- Division of Nephrology, Department of Internal Medicine, Hsin-Jen Hospital, 395 Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 242, Taiwan.
| | - Ming-Cheng Wang
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Shengli Rd., North Dist, Tainan, 704, Taiwan. .,Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
18
|
Atkinson EG, Delgado‐Calle J. The Emerging Role of Osteocytes in Cancer in Bone. JBMR Plus 2019; 3:e10186. [PMID: 30918922 PMCID: PMC6419608 DOI: 10.1002/jbm4.10186] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/29/2019] [Accepted: 02/06/2019] [Indexed: 12/13/2022] Open
Abstract
Advances in the last decade have established the osteocyte, the most abundant cell in bone, as a dynamic and multifunctional cell capable of controlling bone homeostasis by regulating the function of both osteoblasts and osteoclasts. In addition, accumulating evidence demonstrates that osteocyte function is altered in several skeletal disorders, and targeting osteocytes and their derived factors improves skeletal health. Despite the remarkable progress in our understanding of osteocyte biology, there has been a paucity of information regarding the role of osteocytes in the progression of cancer in bone. Exciting, recent discoveries suggest that tumor cells communicate with osteocytes to generate a microenvironment that supports the growth and survival of cancer cells and stimulates bone destruction. This review features these novel findings and discussions regarding the impact of chemotherapy on osteocyte function and the potential of targeting osteocytes for the treatment of cancer in bone. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Emily G Atkinson
- Department of Anatomy and Cell BiologyIndiana University School of MedicineIndianapolisINUSA
| | - Jesús Delgado‐Calle
- Department of Anatomy and Cell BiologyIndiana University School of MedicineIndianapolisINUSA
- Department of MedicineDivision of Hematology/OncologyIndiana University School of MedicineIndianapolisINUSA
- Indiana Center for Musculoskeletal HealthIndiana University School of MedicineIndianapolisINUSA
| |
Collapse
|
19
|
Drake MT, Fenske JS, Blocki FA, Zierold C, Appelman-Dijkstra N, Papapoulos S, Khosla S. Validation of a novel, rapid, high precision sclerostin assay not confounded by sclerostin fragments. Bone 2018; 111:36-43. [PMID: 29550267 PMCID: PMC5924723 DOI: 10.1016/j.bone.2018.03.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/13/2018] [Accepted: 03/13/2018] [Indexed: 10/17/2022]
Abstract
Sclerostin is a 190 amino acid protein secreted primarily by osteocytes. It was initially identified due to mutations in the SOST gene associated with high bone mass phenotypes. Much recent work has sought to determine the importance of sclerostin across an array of conditions which affect the human skeleton. However, accurate measurement of sclerostin from serum and plasma sources remains a significant impediment, with currently available commercial assays showing marked differences in measured sclerostin values. Accordingly, sclerostin assay standardization remains an important but unmet need before sclerostin measurements can be used for the clinical management of bone disease. Here we characterize a novel automated chemiluminescent sclerostin assay (LIAISON®, DiaSorin) which overcomes many of these limitations. Important assay characteristics include: a wide dynamic range (50-6500pg/mL); high intra- (<2.5%) and inter- (<5%) assay precision; matched serum and plasma equivalence (<10% difference); specificity for the intact sclerostin molecule; and rapid assay results. Serum sclerostin levels measured with the LIAISON® assay in a population-based sample of adult men (n=278) and women (n=348) demonstrated that sclerostin levels were significantly higher in men as compared to women and were positively associated with age in both sexes, consistent with previously published work. In postmenopausal women, serum sclerostin levels measured with the LIAISON® assay were reduced in response to treatment with either estrogen or teriparatide, again consistent with previous findings. Collectively, the above data demonstrate that the LIAISON® sclerostin assay provides a reliable tool for more confident assessment of emergent mechanisms wherein sclerostin may impact a number of bone related pathologies.
Collapse
Affiliation(s)
- Matthew T Drake
- Department of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | | | - Frank A Blocki
- DiaSorin Inc., 1951 Northwestern Ave, Stillwater, MN, USA
| | | | | | - Socrates Papapoulos
- Center for Bone Quality, Leiden University Medical Center, Leiden, Netherlands
| | - Sundeep Khosla
- Department of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
20
|
Nicol L, Wang Y, Smith R, Sloan J, Nagamani SC, Shapiro J, Lee B, Orwoll E. Serum Sclerostin Levels in Adults With Osteogenesis Imperfecta: Comparison With Normal Individuals and Response to Teriparatide Therapy. J Bone Miner Res 2018; 33:307-315. [PMID: 29044725 DOI: 10.1002/jbmr.3312] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/10/2017] [Accepted: 10/16/2017] [Indexed: 11/12/2022]
Abstract
Sclerostin (SOST), a glycoprotein primarily derived from osteocytes, is an important regulator of bone remodeling. Osteogenesis imperfecta (OI) is a heritable disorder of bone characterized by low bone mass, bone fragility, recurrent fractures, and bone deformities. Altered SOST-mediated signaling may have a role in pathogenesis of type I collagen-related OI; however, this has not been evaluated in humans. We measured serum SOST levels in adults with OI who were enrolled in a randomized, placebo-controlled clinical trial that evaluated the effects of osteoanabolic therapy with teriparatide. Compared with age- and sex-matched control participants, mean SOST levels were lower in those with type I or types III/VI OI (p < 0.0001). Receiver operating curve analysis revealed that sclerostin alone or sclerostin plus bone mineral content discriminated patients with OI from controls (area under the curve 0.80 and 0.87, respectively). SOST levels increased in the group of patients with type I OI during therapy with teriparatide (compared with placebo, p = 0.01). The increase was significant at 6, 12, and 24 months of therapy (p ≤ 0.02) and was apparent as early as 3 months (p = 0.06). The magnitude of increases in SOST levels during therapy was inversely correlated with increases in vertebral volumetric bone mineral density (vBMD). Overall, these results suggest that: 1) SOST regulation is fundamentally altered in osteogenesis imperfecta; 2) serum SOST levels could be a biomarker of OI in adults; and 3) alterations in SOST may help predict the response to anabolic therapies in OI. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Lindsey Nicol
- Department of Pediatrics, Division of Endocrinology, Oregon Health & Sciences University, Portland, OR, USA
| | - Ying Wang
- Department of Medicine, Division of Biostatistics, Oregon Health & Sciences University, Portland, OR, USA
| | | | - John Sloan
- Lilly Research Laboratories, Indianapolis, IN, USA
| | - Sandesh Cs Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Texas Children's Hospital, Houston, TX, USA
| | - Jay Shapiro
- Department of Bone and Osteogenesis Imperfecta, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Texas Children's Hospital, Houston, TX, USA
| | - Eric Orwoll
- Department of Medicine, Division of Endocrinology, Oregon Health & Sciences University, Portland, OR, USA
| |
Collapse
|
21
|
Delanaye P, Paquot F, Bouquegneau A, Blocki F, Krzesinski JM, Evenepoel P, Pottel H, Cavalier E. Sclerostin and chronic kidney disease: the assay impacts what we (thought to) know. Nephrol Dial Transplant 2017; 33:1404-1410. [DOI: 10.1093/ndt/gfx282] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/23/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
- Pierre Delanaye
- Department of Nephrology, Dialysis, Hypertension, Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
| | - François Paquot
- Department of Nephrology, Dialysis, Hypertension, Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
| | - Antoine Bouquegneau
- Department of Nephrology, Dialysis, Hypertension, Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
| | - Frank Blocki
- Medical LIAISON Bone & Mineral, DiaSorin Inc, Stillwater, MN, USA
| | - Jean-Marie Krzesinski
- Department of Nephrology, Dialysis, Hypertension, Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
| | - Pieter Evenepoel
- Department of Nephrology and Renal Transplantation, University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Hans Pottel
- Department of Public Health and Primary Care at Kulak, University of Leuven, Kortrijk, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, CHU Sart Tilman, Liège, Belgium
| |
Collapse
|
22
|
Circulating levels of sclerostin but not DKK1 associate with laboratory parameters of CKD-MBD. PLoS One 2017; 12:e0176411. [PMID: 28493902 PMCID: PMC5426702 DOI: 10.1371/journal.pone.0176411] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/09/2017] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Mounting evidence indicates that a disturbed Wnt-β-catenin signaling may be involved in the pathogenesis of chronic kidney disease-mineral and bone and mineral disorder (CKD-MBD). Data on the impact of CKD on circulating levels of the Wnt antagonists sclerostin and Dickkopf related protein 1 (DKK1) and the relationship with laboratory parameters of CKD-MBD are incomplete. METHODS We analyzed serum sclerostin and DKK1 in 308 patients across the stages of chronic kidney disease (kDOQI stage 1-2 n = 41; CKD stage 3 n = 54; CKD stage 4-5 n = 54; hemodialysis n = 100; peritoneal dialysis n = 59) as well as in 49 healthy controls. We investigated associations with demographics, renal function, parameters of mineral metabolism including 25(OH) vitamin D, 1,25(OH)2 vitamin D, biointact fibroblast growth factor 23 (FGF23), and parathyroid hormone (PTH), and bone turnover markers. RESULTS Serum sclerostin, but not DKK1, increases in more advanced stages of CKD and associates with PTH, phosphate, and 1,25(OH)2 vitamin D concentrations. Bone turnover markers are highest in hemodialysis patients presenting the combination of high PTH with low sclerostin level. Serum DKK1 levels are lower in CKD patients than in controls and are not associated with laboratory parameters of mineral metabolism. Interestingly, a direct association between DKK1 and platelet count was observed. CONCLUSION In CKD, serum levels of the Wnt inhibitors DKK1 and sclerostin are unrelated, indicating different sites of origin and/ or different regulatory mechanisms. Sclerostin, as opposed to DKK1, may qualify as a biomarker of CKD-MBD, particularly in dialysis patients. DKK1 serum levels, remarkably, correlate almost uniquely with blood platelet counts.
Collapse
|
23
|
Abstract
The Wnt/β-catenin signaling pathway plays an essential role in osteoblast biology. Sclerostin is a soluble antagonist of Wnt/β-catenin signaling secreted primarily by osteocytes. Current evidence indicates that sclerostin likely functions as a local/paracrine regulator of bone metabolism rather than as an endocrine hormone. Nonetheless, circulating sclerostin levels in humans often reflect changes in the bone microenvironment, although there may be exceptions to this observation. Using existing assays, circulating sclerostin levels have been shown to be altered in response to both hormonal stimuli and across a variety of normal physiological and pathophysiological conditions. In both rodents and humans, parathyroid hormone provided either intermittently or continuously suppresses sclerostin levels. Likewise, most evidence from both human and animal studies supports a suppressive effect of estrogen on sclerostin levels. Efforts to examine non-hormonal/systemic regulation of sclerostin have in general shown less consistent findings or have provided associations rather than direct interventional information, with the exception of mechanosensory studies which have consistently demonstrated increased sclerostin levels with skeletal unloading, and conversely decreases in sclerostin with enhanced skeletal loading. Herein, we will review the existent literature on both hormonal and non-hormonal/systemic factors which have been studied for their impact on sclerostin regulation.
Collapse
Affiliation(s)
- Matthew T Drake
- Department of Endocrinology, Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | - Sundeep Khosla
- Department of Endocrinology, Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
24
|
Costa AG, Cremers S, Bilezikian JP. Sclerostin measurement in human disease: Validity and current limitations. Bone 2017; 96:24-28. [PMID: 27742501 DOI: 10.1016/j.bone.2016.10.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/06/2016] [Accepted: 10/10/2016] [Indexed: 10/20/2022]
Abstract
Sclerostin a potent regulator of bone formation, is an antagonist of the Wnt-signaling pathway. The advent of assays to measure circulating sclerostin has enabled research to be performed with the aim to understand the potential role of circulating sclerostin as a pathophysiological marker in a variety of clinical settings. At this time, however, assays to measure circulating sclerostin are still relatively new and have not demonstrated consistent internal agreement in addition to which there are differences between serum and plasma levels. Nevertheless, measurement of sclerostin in the circulation has the potential to reflect the dynamics of bone formation with particular reference to situations in which osteocytes, the major source of circulating sclerostin, may be perturbed. Because of technical uncertainties regarding sclerostin assays that are currently available, circulating sclerostin measurements should be interpreted cautiously with attention to reference ranges for each assay and whether or not the measurement is made in serum or plasma.
Collapse
Affiliation(s)
- Aline G Costa
- Department of Medicine, Division of Endocrinology, Metabolic Bone Diseases Unit, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Serge Cremers
- Department of Medicine, Division of Endocrinology, Metabolic Bone Diseases Unit, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Department of Pathology & Cell Biology, Division of Clinical Pathology College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - John P Bilezikian
- Department of Medicine, Division of Endocrinology, Metabolic Bone Diseases Unit, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| |
Collapse
|
25
|
Decreased Circulating Sclerostin Levels in Renal Transplant Recipients With Persistent Hyperparathyroidism. Transplantation 2016; 100:2188-93. [DOI: 10.1097/tp.0000000000001311] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
26
|
Guañabens N, Ruiz-Gaspà S, Gifre L, Miquel R, Peris P, Monegal A, Dubrueil M, Arias A, Parés A. Sclerostin Expression in Bile Ducts of Patients With Chronic Cholestasis May Influence the Bone Disease in Primary Biliary Cirrhosis. J Bone Miner Res 2016; 31:1725-33. [PMID: 27019303 DOI: 10.1002/jbmr.2845] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/12/2016] [Accepted: 03/23/2016] [Indexed: 02/06/2023]
Abstract
Sclerostin is involved in the regulation of osteoblastogenesis and little is known about its role in the development of bone disease in primary biliary cirrhosis (PBC), characterized by low bone formation. Therefore, we have assessed the circulating levels and the liver expression of sclerostin in this cholestatic disease. Serum sclerostin levels were measured in 79 women with PBC (mean age 60.6 ± 1.2 years) and in 80 control women. Lumbar and femoral bone mineral density (BMD), as well as parameters of mineral metabolism and bone remodeling, were measured. Moreover, sclerostin gene (SOST) expression in the liver was assessed by real-time PCR in samples of liver tissue taken by biopsy in 11 PBC patients and in 5 normal liver specimens. Presence and distribution of sclerostin was evaluated in liver slices from 11 patients by immunohistochemistry. The severity of histologic lesions was assessed semiquantitatively in the same liver samples. PBC patients had higher sclerostin levels than controls (75.6 ± 3.9 versus 31.7 ± 1.6 pmol/L, p < 0.001). Serum sclerostin correlated inversely with markers of bone formation and resorption. Sclerostin mRNA in the liver was overexpressed compared with control samples (2.7-fold versus healthy liver). Sclerostin was detected by immunohistochemistry in 7 of the 11 liver samples, mainly located in the bile ducts. Liver sclerostin was associated with the severity of cholangitis (p = 0.02) and indirectly with the degree of lobular inflammation (p = 0.03). Sclerostin mRNA expression was higher in samples that tested positive by immunohistochemistry and particularly in those with lobular granuloma (p = 0.02). The increased expression of sclerostin in the liver and the association with histologic cholangitis may explain the high serum levels of this protein in patients with PBC, thus suggesting that sclerostin may influence the decreased bone formation in this cholestatic disease. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Núria Guañabens
- Department of Rheumatology, Metabolic Bone Diseases Unit, Hospital Clínic, Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
- Center for Biomedical Research Network in Hepatic and Digestive Diseases (CIBERehd), Hospital Clínic, Barcelona, Spain
| | - Silvia Ruiz-Gaspà
- Center for Biomedical Research Network in Hepatic and Digestive Diseases (CIBERehd), Hospital Clínic, Barcelona, Spain
| | - Laia Gifre
- Department of Rheumatology, Metabolic Bone Diseases Unit, Hospital Clínic, Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Rosa Miquel
- Pathology Department, Hospital Clínic, Barcelona, Spain
| | - Pilar Peris
- Department of Rheumatology, Metabolic Bone Diseases Unit, Hospital Clínic, Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
- Center for Biomedical Research Network in Hepatic and Digestive Diseases (CIBERehd), Hospital Clínic, Barcelona, Spain
| | - Ana Monegal
- Department of Rheumatology, Metabolic Bone Diseases Unit, Hospital Clínic, Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Marta Dubrueil
- Center for Biomedical Research Network in Hepatic and Digestive Diseases (CIBERehd), Hospital Clínic, Barcelona, Spain
| | - Ana Arias
- Department of Rheumatology, Metabolic Bone Diseases Unit, Hospital Clínic, Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Albert Parés
- Center for Biomedical Research Network in Hepatic and Digestive Diseases (CIBERehd), Hospital Clínic, Barcelona, Spain
- Liver Unit, Hospital Clínic, Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| |
Collapse
|
27
|
Piec I, Washbourne C, Tang J, Fisher E, Greeves J, Jackson S, Fraser WD. How Accurate is Your Sclerostin Measurement? Comparison Between Three Commercially Available Sclerostin ELISA Kits. Calcif Tissue Int 2016; 98:546-55. [PMID: 26749312 PMCID: PMC4860200 DOI: 10.1007/s00223-015-0105-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/30/2015] [Indexed: 12/30/2022]
Abstract
Sclerostin, bone formation antagonist is in the spotlight as a potential biomarker for diseases presenting with associated bone disorders such as chronic kidney disease (CDK-MBD). Accurate measurement of sclerostin is therefore important. Several immunoassays are available to measure sclerostin in serum and plasma. We compared the performance of three commercial ELISA kits. We measured sclerostin concentrations in serum and EDTA plasma obtained from healthy young (18-26 years) human subjects using kits from Biomedica, TECOmedical and from R&D Systems. The circulating sclerostin concentrations were systematically higher when measured with the Biomedica assay (serum: 35.5 ± 1.1 pmol/L; EDTA: 39.4 ± 2.0 pmol/L; mean ± SD) as compared with TECOmedical (serum: 21.8 ± 0.7 pmol/L; EDTA: 27.2 ± 1.3 pmol/L) and R&D Systems (serum: 7.6 ± 0.3 pmol/L; EDTA: 30.9 ± 1.5 pmol/L). We found a good correlation between the assay for EDTA plasma (r > 0.6; p < 0.001) while in serum, only measurements obtained using TECOmedical and R&D Systems assays correlated significantly (r = 0.78; p < 0.001). There was no correlation between matrices results when using the Biomedica kit (r = 0.20). The variability in values generated from Biomedica, R&D Systems and TECOmedical assays raises questions regarding the accuracy and specificity of the assays. Direct comparison of studies using different kits is not possible and great care should be given to measurement of sclerostin, with traceability of reagents. Standardization with appropriate material is required before different sclerostin assays can be introduced in clinical practice.
Collapse
Affiliation(s)
- Isabelle Piec
- Bioanalytical Facility, University of East Anglia, Floor 2, Bob Champion Research and Education Building, Norwich Research Park, James Watson Road, Norwich, NR4 7UQ, UK.
| | - Christopher Washbourne
- Bioanalytical Facility, University of East Anglia, Floor 2, Bob Champion Research and Education Building, Norwich Research Park, James Watson Road, Norwich, NR4 7UQ, UK
| | - Jonathan Tang
- Bioanalytical Facility, University of East Anglia, Floor 2, Bob Champion Research and Education Building, Norwich Research Park, James Watson Road, Norwich, NR4 7UQ, UK
| | - Emily Fisher
- Bioanalytical Facility, University of East Anglia, Floor 2, Bob Champion Research and Education Building, Norwich Research Park, James Watson Road, Norwich, NR4 7UQ, UK
| | - Julie Greeves
- Women Ground Close Combat Review, Directorate of Manning (Army), Blenheim Bld, IDL 27, Monxton Road, Andover, SP11 8HT, UK
| | - Sarah Jackson
- Women Ground Close Combat Review, Directorate of Manning (Army), Blenheim Bld, IDL 27, Monxton Road, Andover, SP11 8HT, UK
| | - William D Fraser
- Bioanalytical Facility, University of East Anglia, Floor 2, Bob Champion Research and Education Building, Norwich Research Park, James Watson Road, Norwich, NR4 7UQ, UK
- Norfolk and Norwich University Hospital, Norwich, NR4 7UV, UK
| |
Collapse
|
28
|
Tsentidis C, Gourgiotis D, Kossiva L, Marmarinos A, Doulgeraki A, Karavanaki K. Sclerostin distribution in children and adolescents with type 1 diabetes mellitus and correlation with bone metabolism and bone mineral density. Pediatr Diabetes 2016; 17:289-99. [PMID: 26094958 DOI: 10.1111/pedi.12288] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 03/28/2015] [Accepted: 05/08/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Sclerostin is an inhibitor of the Wnt/beta-catenin bone metabolic pathway. Increased sclerostin levels and reduced bone mineral density (BMD) have been documented in adult patients with diabetes mellitus (DM), predominantly in those with type 2 diabetes mellitus (T2DM). No relative data exist on childhood type 1 diabetes mellitus (T1DM). Our objective was to study plasma sclerostin in T1DM children and adolescents and controls and its correlations with metabolic bone markers and BMD. SUBJECTS AND METHODS This was a cross-sectional study that was conducted at an outpatient clinical center. Forty T1DM children and adolescents were evaluated (mean ± SD age: 13.04 ± 3.53 yr, T1DM duration: 5.15 ± 3.33 yr), along with 40 healthy matched controls (age 12.99 ± 3.3 yr). Sclerostin, soluble receptor activator of nuclear factor-kappaB ligand (s-RANKL), osteoprotegerin, osteocalcin, C-telopeptide crosslinks, electrolytes, parathyroid hormone (PTH), and total 25(OH)D were measured. Lumbar and subcranial total body BMD were evaluated with dual energy X-ray absorptiometry (DXA). RESULTS Sclerostin levels demonstrated a Gaussian distribution, with no significant difference between patients and controls (51.56 ± 12.05 vs. 50.98 ± 13.55 pmol/L, p = 0.84). Significantly lower values were found in girls and prepubertal children. Sclerostin values were significantly and gradually increased in children through pubertal Tanner stages 1-3, were reduced at stage 4 and increased again at pubertal stage 5. Sclerostin levels were positively correlated with logCTX (logarithm of C-terminal telopeptide crosslinks of type I collagen), logOsteocalcin (logarithm of Osteocalcin), magnesium, total body, and L1-L4 BMD z-score. CONCLUSIONS T1DM patients had similar levels of sclerostin with controls. Sclerostin correlated with bone resorption and formation markers and also with bone mass indices, gender, and pubertal stage. The decrease in sclerostin values observed in pubertal stage 4 adolescents coincides with the concurrent growth spurt, and is consistent with sclerostin physiology as an inhibiting signal.
Collapse
Affiliation(s)
- Charalampos Tsentidis
- Diabetic Clinic, 2nd Department of Pediatrics, Athens University Medical School, 'P&A Kyriakou' Children's Hospital, Athens, Greece
| | - Dimitrios Gourgiotis
- Laboratory of Clinical Biochemistry - Molecular Diagnostics, 2nd Department of Pediatrics, Athens University Medical School, 'P & A Kyriakou' Children's Hospital, Athens, Greece
| | - Lydia Kossiva
- Diabetic Clinic, 2nd Department of Pediatrics, Athens University Medical School, 'P&A Kyriakou' Children's Hospital, Athens, Greece
| | - Antonios Marmarinos
- Laboratory of Clinical Biochemistry - Molecular Diagnostics, 2nd Department of Pediatrics, Athens University Medical School, 'P & A Kyriakou' Children's Hospital, Athens, Greece
| | - Artemis Doulgeraki
- Department of Bone and Mineral Metabolism, Institute of Child Health, 'Aghia Sophia' Children's Hospital, Athens, Greece
| | - Kyriaki Karavanaki
- Diabetic Clinic, 2nd Department of Pediatrics, Athens University Medical School, 'P&A Kyriakou' Children's Hospital, Athens, Greece
| |
Collapse
|
29
|
Abstract
For many years, osteocytes have been the forgotten bone cells and considered as inactive spectators buried in the bone matrix. We now know that osteocytes detect and respond to mechanical and hormonal stimuli to coordinate bone resorption and bone formation. Osteocytes are currently considered a major source of molecules that regulate the activity of osteoclasts and osteoblasts, such as RANKL and sclerostin; and genetic and pharmacological manipulations of either molecule markedly affect bone homeostasis. Besides playing a role in physiological bone homeostasis, accumulating evidence supports the notion that dysregulation of osteocyte function and alteration of osteocyte life-span underlies the pathophysiology of skeletal disorders characterized by loss bone mass and increased bone fragility, as well as the damaging effects of cancer in bone. In this review, we highlight some of these investigations and discuss novel observations that demonstrate that osteocytes, far from being passive cells entombed in the bone, are critical for bone function and maintenance.
Collapse
Affiliation(s)
- Jesus Delgado-Calle
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana ; Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana ; Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, Indiana ; Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| |
Collapse
|
30
|
Farr JN, Roforth MM, Fujita K, Nicks KM, Cunningham JM, Atkinson EJ, Therneau TM, McCready LK, Peterson JM, Drake MT, Monroe DG, Khosla S. Effects of Age and Estrogen on Skeletal Gene Expression in Humans as Assessed by RNA Sequencing. PLoS One 2015; 10:e0138347. [PMID: 26402159 PMCID: PMC4581624 DOI: 10.1371/journal.pone.0138347] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 08/26/2015] [Indexed: 01/19/2023] Open
Abstract
UNLABELLED Precise delineation of the specific genes and pathways altered with aging and estrogen (E) therapy may lead to new skeletal biomarkers and the development of novel bone therapeutics. Previous human bone studies, however, have been limited by only examining pre-specified genes and pathways. High-throughput RNA sequencing (RNAseq), on the other hand, offers an unbiased approach to examine the entire transcriptome. Here we present an RNAseq analysis of human bone samples, obtained from iliac crest needle biopsies, to yield the first in vivo interrogation of all genes and pathways that may be altered in bone with aging and E therapy in humans. 58 healthy women were studied, including 19 young women (mean age ± SD, 30.3 ± 5.4 years), 19 old women (73.1 ± 6.6 years), and 20 old women treated with 3 weeks of E therapy (70.5 ± 5.2 years). Using generally accepted criteria (false discovery rate [q] < 0.10), aging altered a total of 678 genes and 12 pathways, including a subset known to regulate bone metabolism (e.g., Notch). Interestingly, the LEF1 transcription factor, which is a classical downstream target of the Wnt/β-catenin signaling pathway, was significantly downregulated in the bones from the old versus young women; consistent with this, LEF1 binding sites were significantly enriched in the promoter regions of the differentially expressed genes in the old versus young women, suggesting that aging was associated with alterations in Wnt signaling in bone. Further, of the 21 unique genes altered in bone by E therapy, the expression of INHBB (encoding for the inhibin, beta B polypeptide), which decreased with aging (by 0.6-fold), was restored to young adult levels in response to E therapy. In conclusion, our data demonstrate that aging alters a substantial portion of the skeletal transcriptome, whereas E therapy appears to have significant, albeit less wide-ranging effects. These data provide a valuable resource for the potential identification of novel biomarkers associated with age-related bone loss and also highlight potential pathways that could be targeted to treat osteoporosis. TRIAL REGISTRATION ClinicalTrials.gov NCT02349113.
Collapse
Affiliation(s)
- Joshua N. Farr
- Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - Matthew M. Roforth
- Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - Koji Fujita
- Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - Kristy M. Nicks
- Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - Julie M. Cunningham
- Department of Experimental Pathology and Laboratory Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - Elizabeth J. Atkinson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - Terry M. Therneau
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - Louise K. McCready
- Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - James M. Peterson
- Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - Matthew T. Drake
- Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - David G. Monroe
- Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
| | - Sundeep Khosla
- Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, 55905, United States of America
- * E-mail:
| |
Collapse
|
31
|
|
32
|
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: 352] [Impact Index Per Article: 35.2] [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
|
33
|
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: 91] [Impact Index Per Article: 9.1] [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
|
34
|
Uremic toxicity and sclerostin in chronic kidney disease patients. Nephrol Ther 2014; 10:463-70. [PMID: 25070604 DOI: 10.1016/j.nephro.2014.04.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 04/10/2014] [Accepted: 04/10/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND AIMS Sclerostin is a circulating inhibitor of the Wnt/β-catenin pathway and may have a role in chronic kidney disease (CKD)-mineral and bone disorder. Blood sclerostin levels are known to be elevated in patients undergoing maintenance dialysis. The aims of the present study were to evaluate sclerostin levels in patients at different CKD stages and study potential associations between sclerostin levels and (i) biochemical parameters that are disturbed in CKD, (ii) markers of vascular disease and (iii) mortality. METHODS One hundred and forty patients at CKD stages 2-5D were included in the present study. Routine clinical biochemistry tests and assays for sclerostin, protein-bound uremic toxins (indoxylsulphate [IS] and p-cresyl sulphate [PCS]) and the toxin β2 microglobulin (β2M) were performed. Aortic and coronary calcification and arterial stiffness were assessed by multislice spiral computed tomography and pulse wave velocity measurements. The enrolled patients were prospectively monitored for mortality. RESULTS Sclerostin levels were found to be elevated in CKD patients (especially those on hemodialysis). Furthermore, sclerostin levels were positively correlated with inflammation markers, phosphate, fibroblast growth factor 23, IS, PCS, β2M and arterial stiffness. A multivariate linear regression analysis indicated that sclerostin levels were independently associated with IS, PCS and β2M levels. Elevated serum sclerostin appeared to be associated with mortality (independently of age and inflammation). However, this association disappeared after adjustment for a propensity score including age, phosphate, interleukin-6, CKD stage and PCS. CONCLUSION Our results indicate that sclerostin levels are elevated in CKD patients and are associated with inflammation, vascular lesions, uremia and (potentially) mortality.
Collapse
|
35
|
Costa AG, Bilezikian JP, Lewiecki EM. Update on romosozumab : a humanized monoclonal antibody to sclerostin. Expert Opin Biol Ther 2014; 14:697-707. [PMID: 24665957 DOI: 10.1517/14712598.2014.895808] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Disorders with inactivating mutations of the SOST gene result in reduced or absent expression of sclerostin and are associated with high bone mass. Sclerostin is an important regulator of bone formation due to its inhibitory actions in the osteoanabolic Wnt signaling pathway. Advances in understanding the mechanisms of action of this signaling molecule have led to the development of a pharmacological inhibitor of sclerostin with potential clinical applications as an osteoanabolic drug for the treatment of osteoporosis. AREAS COVERED Romosozumab is the first humanized monoclonal sclerostin antibody to be tested in clinical trials. Similar to preclinical animal studies with sclerostin antibodies, initial clinical studies show that romosozumab increases bone formation and bone mineral density. EXPERT OPINION Blocking sclerostin action with romosozumab is a promising new therapeutic approach to osteoanabolic therapy of osteoporosis; efficacy and safety data on large controlled studies are awaited.
Collapse
Affiliation(s)
- Aline G Costa
- Columbia University, College of Physicians and Surgeons, Division of Endocrinology, Department of Medicine, Metabolic Bone Diseases Unit , 630 West 168th Street, NY 10032 , USA
| | | | | |
Collapse
|
36
|
Al-Zifzaf DS, Hamza SA, Kaddah EA, Abo-Shady RA. Type 2 diabetes raises serum sclerostin levels and disturbs the relation between sclerostin and bone mineral density: a call for caution with antisclerostin therapy in osteoporosis. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2014. [DOI: 10.4103/1110-161x.128136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
37
|
Klangjareonchai T, Nimitphong H, Saetung S, Bhirommuang N, Samittarucksa R, Chanprasertyothin S, Sudatip R, Ongphiphadhanakul B. Circulating sclerostin and irisin are related and interact with gender to influence adiposity in adults with prediabetes. Int J Endocrinol 2014; 2014:261545. [PMID: 25276128 PMCID: PMC4167818 DOI: 10.1155/2014/261545] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/29/2014] [Accepted: 08/17/2014] [Indexed: 11/17/2022] Open
Abstract
Objectives. Sclerostin, an osteocyte-specific protein, has been found to be related to adiposity and glucose metabolism. Irisin, a myokine, can affect browning of white fat and influence glucose and energy homeostasis. Taken together, this suggests a probable network among fat, bone, and muscle that may influence health outcomes. The aims of this study were to investigate the relationship of circulating sclerostin and irisin and their association with adiposity (assessed by body mass index (BMI)). Materials/Methods. A cross-sectional study included 98 adults with impaired fasting glucose and/or impaired glucose tolerance. 75 gm OGTT was performed in all subjects. Fasting plasma samples were obtained for glycated hemoglobin, calcium, creatinine, serum sclerostin and irisin. Results. Circulating irisin and sclerostin were highly correlated (r = -0.4; P < 0.001). After controlling for age, gender, and BMI, irisin was significantly related to sclerostin (P < 0.001). Multivariate linear regression analysis demonstrated that circulating sclerostin (β = -0.45; P < 0.05) and irisin (β = -0.46; P < 0.05) were negatively associated with BMI, independent of age in males. In females, no relationship of sclerostin or irisin to BMI was found. Conclusions. Circulating irisin and sclerostin are highly related. Interventions targeting irisin could affect sclerostin and vice versa.
Collapse
Affiliation(s)
- Theerawut Klangjareonchai
- Department of Medicine and Research Center, Ramathibodi Hospital, Mahidol University, Rama VI Street, Rajathevi, Bangkok 10400, Thailand
| | - Hataikarn Nimitphong
- Department of Medicine and Research Center, Ramathibodi Hospital, Mahidol University, Rama VI Street, Rajathevi, Bangkok 10400, Thailand
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Rama 6 Road, Rajathevi, Bangkok 10400, Thailand
- *Hataikarn Nimitphong:
| | - Sunee Saetung
- Department of Medicine and Research Center, Ramathibodi Hospital, Mahidol University, Rama VI Street, Rajathevi, Bangkok 10400, Thailand
| | - Nuttapimon Bhirommuang
- Department of Medicine and Research Center, Ramathibodi Hospital, Mahidol University, Rama VI Street, Rajathevi, Bangkok 10400, Thailand
| | - Rattanapan Samittarucksa
- Department of Medicine and Research Center, Ramathibodi Hospital, Mahidol University, Rama VI Street, Rajathevi, Bangkok 10400, Thailand
| | - Suwannee Chanprasertyothin
- Department of Medicine and Research Center, Ramathibodi Hospital, Mahidol University, Rama VI Street, Rajathevi, Bangkok 10400, Thailand
| | - Rattana Sudatip
- Department of Medicine and Research Center, Ramathibodi Hospital, Mahidol University, Rama VI Street, Rajathevi, Bangkok 10400, Thailand
| | - Boonsong Ongphiphadhanakul
- Department of Medicine and Research Center, Ramathibodi Hospital, Mahidol University, Rama VI Street, Rajathevi, Bangkok 10400, Thailand
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Rama 6 Road, Rajathevi, Bangkok 10400, Thailand
| |
Collapse
|
38
|
Abstract
Osteocytes, the most abundant cells in bone, have been long postulated to detect and respond to mechanical and hormonal stimuli and to coordinate the function of osteoblasts and osteoclasts. The discovery that the inhibitor of bone formation sclerostin is primarily expressed in osteocytes in bone and downregulated by anabolic stimuli provided a mechanism by which osteocytes influence the activity of osteoblasts. Advances of the last few years provided experimental evidence demonstrating that osteocytes also participate in the recruitment of osteoclasts and the initiation of bone remodeling. Apoptotic osteocytes trigger yet-to-be-identified signals that attract osteoclast precursors to specific areas of bone, which in turn differentiate to mature, bone-resorbing osteoclasts. Osteocytes are also the source of molecules that regulate the generation and activity of osteoclasts, such as OPG and RANKL; and genetic manipulations of the mouse genome leading to loss or gain of function or to altered expression of either molecule in osteocytes markedly affect bone resorption. This review highlights these investigations and discusses how the novel concept of osteocyte-driven bone resorption and formation impacts our understanding of the mechanisms by which current therapies control bone remodeling.
Collapse
Affiliation(s)
- Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS5035, Indianapolis, IN, 46202, USA,
| |
Collapse
|
39
|
Kim SH, Yoon SY, Lim SK, Rhee Y. The effect of renal dysfunction on circulating sclerostin level in patients with type 2 diabetes. Int J Endocrinol 2014; 2014:715908. [PMID: 25053944 PMCID: PMC4099222 DOI: 10.1155/2014/715908] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/07/2014] [Accepted: 06/10/2014] [Indexed: 11/17/2022] Open
Abstract
Objective. Sclerostin is a Wnt inhibitor produced specifically by osteocytes. However, it is not currently clear whether renal dysfunction has an effect on circulating sclerostin level in patients with type 2 diabetes. The aim of the study was to evaluate this relationship. Design and Patients. We conducted a cross-sectional observational study of 302 type 2 diabetic patients with or without chronic kidney disease. Serum sclerostin level was analyzed by ELISA, and renal function was assessed by estimated glomerular filtration rate (eGFR) using chronic kidney disease epidemiology collaboration (CKD-EPI) equation. Results. There was a strong correlation between sclerostin level with renal function presented as serum creatinine (r = 0.745, P < 0.001) and eGFR (r = -0.590, P < 0.001). Serum sclerostin level was significantly higher in patients with CKD-G3 stage than those with CKD-G1/2 stages after adjusting for age, sex, and BMI (P = 0.011). Patients with CKD-G4/5 stages had dramatically increased level of circulating sclerostin. Multiple regression analyses found that age, sex, and eGFR were independent determining factors for circulating sclerostin level. Conclusion. Our data showed that serum sclerostin levels start to increase in diabetic patients with CKD-G3 stage. Further studies are needed to establish the potential role of elevated sclerostin in diabetic patients with CKD.
Collapse
Affiliation(s)
- Se Hwa Kim
- Division of Endocrinology, Department of Internal Medicine, Kwandong University College of Medicine, 100-25 Simgok-ro, Seo-gu, Incheon 404-834, Republic of Korea
| | - Soo Young Yoon
- Division of Nephrology, Department of Internal Medicine, Kwandong University College of Medicine, 100-25 Simgok-ro, Seo-gu, Incheon 404-834, Republic of Korea
| | - Sung-Kil Lim
- Department of Internal Medicine, Severance Hospital, Endocrine Research Institute, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 120-752, Republic of Korea
| | - Yumie Rhee
- Department of Internal Medicine, Severance Hospital, Endocrine Research Institute, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 120-752, Republic of Korea
| |
Collapse
|
40
|
Costa AG, Walker MD, Zhang CA, Cremers S, Dworakowski E, McMahon DJ, Liu G, Bilezikian JP. Circulating sclerostin levels and markers of bone turnover in Chinese-American and white women. J Clin Endocrinol Metab 2013; 98:4736-43. [PMID: 24037879 PMCID: PMC3849675 DOI: 10.1210/jc.2013-2106] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Chinese-American women have bone microarchitectural features that confer greater bone stiffness compared to white women, but the physiology underlying these findings has not been investigated. OBJECTIVE The purpose of the study was to assess racial differences in serum sclerostin and bone turnover markers (BTMs), and to explore their associations with each other, volumetric bone mineral density (BMD), and bone microarchitecture in Chinese-American and white women. DESIGN AND SETTING We conducted a cross-sectional study at a university hospital. PARTICIPANTS We studied 138 women. RESULTS Serum osteocalcin was 19-28% lower in pre- and postmenopausal Chinese-American vs white women, respectively (both P < .01). C-Terminal telopeptide of type I collagen (CTX) level was 18-22% lower in pre- and postmenopausal Chinese-American vs white women (both P < .05). Pre- vs postmenopausal differences in osteocalcin and CTX were greater in white vs Chinese-American women. Sclerostin levels were similar in both races, but BTMs were differentially associated with sclerostin by race and menopausal status. BTMs were not correlated with sclerostin in Chinese-Americans. CTX and bone-specific alkaline phosphatase were positively associated with sclerostin (r = 0.353, r = 0.458; both P < .05) in white premenopausal women. In contrast, in postmenopausal white women, the associations of sclerostin with amino-terminal propeptide of type I procollagen, isoform 5b of tartrate-resistant acid phosphatase, and CTX were negative (all P < .05). Adjusting for covariates, sclerostin was positively associated with areal BMD in both races. CONCLUSIONS Lower BTMs in Chinese-American women and greater age-related differences in BTMs among white women provide a physiological framework to account for racial differences in BMD, microarchitecture, and fracture.
Collapse
Affiliation(s)
- Aline G Costa
- 630 West 168th Street, PH8 West-864, New York, New York 10032.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhou YJ, Li A, Song YL, Zhou H, Li Y, Tang YS. Role of sclerostin in the bone loss of postmenopausal chinese women with type 2 diabetes. ACTA ACUST UNITED AC 2013; 28:135-9. [PMID: 24074614 DOI: 10.1016/s1001-9294(13)60038-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To evaluate the role of sclerostin in bone loss of postmenopausal Chinese women with type 2 diabetes mellitus. METHODS The postmenopausal patients suffering from type 2 diabetes mellitus and age, body mass index, and duration of menopause matched healthy controls were enrolled into this cross-sectional study according to criteria of inclusion and exclusion. The serum sclerostin level and bone mineral density of the anterior-posterior lumbar spine (L1-L4), femoral neck, and total hip were determined by using a quantitative sandwich ELISA kit and dual X-ray absorptiometry, respectively. Meanwhile, the clinical and laboratory indexes of bone mineral metabolism were analyzed. Associations between serum sclerostin level and bone mineral density as well as bone turnover markers were evaluated by linear regression analysis. RESULTS Finally, 265 postmenopausal women with type 2 diabetes and 225 non-diabetic women were recruited in the diabetic group and control group, respectively. Serum sclerostin level of the diabetic group was significantly higher than that of the control group (48.2±19.4 vs. 37.2±18.6 pmol/L, P<0.001) and was increased with age in both groups (diabetic group, r=0.374, P<0.001; control group, r=0.312, P<0.001). In type 2 diabetes patients, serum sclerostin concentration was positively correlated with hemoglobin A1c level (r=0.237; P=0.021). Biochemical bone turnover markers, intact parathyroid hormone and bone-specific alkaline phosphatase, were negatively associated with serum sclerostin level (r=-0.138, P=0.078 and r=-0.265, P<0.001). Conversely, the positive correlation between sclerostin and C-terminal cross-linking telopeptide of type I collagen was found in diabetic patients (r=0.354, P<0.001). Serum sclerostin levels of the diabetic group were positively correlated with bone mineral density of the lumbar spine, femoral neck, and total hip (r=0.324, 0.367, and 0.416, respectively; all P<0.001). CONCLUSIONS Sclerostin might participate in the pathogenesis of bone loss of type 2 diabetes. The high sclerostin level might serve as a marker of increased osteocyte activity in postmenopausal patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Yi-jun Zhou
- Department of Endocrinology and Metabolism, Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Osteoporosis is a skeletal disorder characterized by bone loss, which results in architectural deterioration of the skeleton, compromised bone strength and an increased risk of fragility fractures. Most current therapies for osteoporosis stabilize the skeleton by inhibiting bone resorption (antiresorptive agents), but the development of anabolic therapies that can increase bone formation and bone mass is of great interest. Wnt signalling induces differentiation of bone-forming cells (osteoblasts) and suppresses the development of bone-resorbing cells (osteoclasts). The Wnt pathway is controlled by antagonists that interact either directly with Wnt proteins or with Wnt co-receptors. The importance of Wnt signalling in bone formation is indicated by skeletal disorders such as sclerosteosis and van Buchem syndrome, which are caused by mutations in the gene encoding the Wnt antagonist sclerostin (SOST). Experiments in mice have shown that downregulation or neutralization of Wnt antagonists enhances bone formation. Phase II clinical trials show that 1-year treatment with antisclerostin antibodies increases bone formation, decreases bone resorption and leads to a substantial increase in BMD. Consequently, Wnt signalling can be targeted by the neutralization of its extracellular antagonists to obtain a skeletal anabolic response.
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Research, Saint Francis Hospital and Medical Centre, 114 Woodland Street, Hartford, CT 06105-1299, USA.
| |
Collapse
|
43
|
Jean G, Chazot C. Sclerostin in CKD-MBD: one more paradoxical bone protein? Nephrol Dial Transplant 2013; 28:2932-5. [DOI: 10.1093/ndt/gft222] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
44
|
Rossini M, Gatti D, Adami S. Involvement of WNT/β-catenin signaling in the treatment of osteoporosis. Calcif Tissue Int 2013; 93:121-32. [PMID: 23748710 DOI: 10.1007/s00223-013-9749-z] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 05/05/2013] [Indexed: 12/17/2022]
Abstract
Osteoblast differentiation is predominantly regulated by the WNT/β-catenin signaling (canonical WNT pathway), which, together with bone morphogenetic proteins, acts as the master regulator of osteogenesis. The recent characterization of the canonical WNT pathway in the regulation of bone modeling and remodeling provided important insights for our understanding of the pathophysiology of a number of conditions and of the mechanism of action of hormones or drugs with important effect on bone metabolism. This review is mainly focused on the growing therapeutic implications of these new findings. WNT/β-catenin signaling plays a key role in bone tissue by determining the differentiation of stem cells into mature osteoblasts rather than into chondrocytes and adipocytes. Its regulation is predominantly driven by the production of two WNT signaling antagonists: sclerostin (SOST) and Dickkopf-related protein 1 (DKK1). The most proximate regulator of SOST expression by osteocytes and its serum levels is bone mechanical load. SOST expression is increased with advancing age, by glucocorticoid treatment and during treatment with antiresorptive agents such as bisphosphonates and denosumab, while it is decreased by parathyroid hormone excess or administration of estrogens. Correlation between DKK1 serum levels and bone formation in various pathological conditions or during osteoporosis treatment has been reported. Inhibitors of the negative regulators of WNT/β-catenin signaling ("inhibiting the endogenous inhibitors") are potential candidates for the prevention and treatment of bone loss. Inactivating monoclonal antibodies against SOST appears to be the most attractive strategy because SOST is the only component of the WNT pathway expressed almost exclusively by osteocytes.
Collapse
Affiliation(s)
- Maurizio Rossini
- Department of Medicine, Rheumatology Section, Policlinico Borgo Roma, University of Verona, Piazzale Scuro, 10, 37134, Verona, Italy
| | | | | |
Collapse
|
45
|
Clinical utility of serum sclerostin measurements. BONEKEY REPORTS 2013; 2:361. [PMID: 24578825 DOI: 10.1038/bonekey.2013.95] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 05/16/2013] [Indexed: 01/16/2023]
Abstract
Sclerostin is an osteocyte-secreted soluble antagonist of the Wnt/β-catenin signaling pathway requisite for osteoblast development and activity. Efforts over the past several years have focused on unraveling the role of sclerostin in both normal physiological and pathological conditions. Sclerostin levels are undetectable in the serum of patients with sclerosteosis. In normal individuals, serum sclerostin levels are higher in males and increase in both sexes across the adult lifespan. Some, but not other, studies have demonstrated that higher serum sclerostin levels are associated with increased fracture risk, particularly when paired with lower bone mineral density. Levels of circulating sclerostin are highly correlated with bone marrow sclerostin levels. Sclerostin levels are inversely related to parathyroid hormone levels. Clinical conditions in which serum sclerostin levels have been measured include ankylosing spondylitis, chronic kidney disease, diabetes, fractures, hypercortisolism, multiple myeloma and spinal cord injury. Even within clearly defined clinical conditions, however, consistent changes in serum sclerostin levels have not always been seen. This may reflect differences in currently available commercial assays or sample sources (serum versus plasma), and suggests further study is needed before sclerostin measurements are introduced into routine clinical practice. Until such issues are resolved, measurement of sclerostin levels appears to be most useful for understanding the mechanisms by which osteocytes regulate bone turnover through the integration of hormonal, physical and pharmacological stimuli, rather than to guide clinical-care decisions.
Collapse
|
46
|
Bellido T, Saini V, Pajevic PD. Effects of PTH on osteocyte function. Bone 2013; 54:250-7. [PMID: 23017659 PMCID: PMC3552098 DOI: 10.1016/j.bone.2012.09.016] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Revised: 08/22/2012] [Accepted: 09/17/2012] [Indexed: 12/11/2022]
Abstract
Osteocytes are ideally positioned to detect and respond to mechanical and hormonal stimuli and to coordinate the function of osteoblasts and osteoclasts. However, evidence supporting the involvement of osteocytes in specific aspects of skeletal biology has been limited mainly due to the lack of suitable experimental approaches. Few crucial advances in the field in the past several years have markedly increased our understanding of the function of osteocytes. The development of osteocytic cell lines initiated a plethora of in vitro studies that have provided insights into the unique biology of osteocytes and continue to generate novel hypotheses. Genetic approaches using promoter fragments that direct gene expression to osteocytes allowed the generation of mice with gain or loss of function of particular genes revealing their role in osteocyte function. Furthermore, evidence that Sost/sclerostin is expressed primarily in osteocytes and inhibits bone formation by osteoblasts, fueled research attempting to identify regulators of this gene as well as other osteocyte products that impact the function of osteoblasts and osteoclasts. The discovery that parathyroid hormone (PTH), a central regulator of bone homeostasis, inhibits sclerostin expression generated a cascade of studies that revealed that osteocytes are crucial target cells of the actions of PTH. This review highlights these investigations and discusses their significance for advancing our understanding of the mechanisms by which osteocytes regulate bone homeostasis and for developing therapies for bone diseases targeting osteocytes.
Collapse
Affiliation(s)
- Teresita Bellido
- Department of Anatomy and Cell Biology
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine
- Corresponding authors and reprint requests: Teresita Bellido, Ph.D., Department of Anatomy and Cell Biology, and Department of Internal Medicine, Division of Endocrinology, Indiana University School of Medicine, 635 Barnhill Drive, MS5035, Indianapolis, IN 46202, . Paola Divieti Pajevic, M.D., Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Thier 1101, 50 Blossom Street, Boston, Massachusetts 02114, USA,
| | - Vaibhav Saini
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School
| | - Paola Divieti Pajevic
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School
- Corresponding authors and reprint requests: Teresita Bellido, Ph.D., Department of Anatomy and Cell Biology, and Department of Internal Medicine, Division of Endocrinology, Indiana University School of Medicine, 635 Barnhill Drive, MS5035, Indianapolis, IN 46202, . Paola Divieti Pajevic, M.D., Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Thier 1101, 50 Blossom Street, Boston, Massachusetts 02114, USA,
| |
Collapse
|
47
|
Yao W, Dai W, Jiang JX, Lane NE. Glucocorticoids and osteocyte autophagy. Bone 2013; 54:279-84. [PMID: 23356984 PMCID: PMC3784314 DOI: 10.1016/j.bone.2013.01.034] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 01/06/2013] [Accepted: 01/18/2013] [Indexed: 12/17/2022]
Abstract
Glucocorticoids are used for the treatment of inflammatory and autoimmune diseases. While they are effective therapy, bone loss and incident fracture risk are high. While previous studies have found GC effects on both osteoclasts and osteoblasts, our work has focused on the effects of GCs on osteocytes. Osteocytes exposed to low dose GCs undergo autophagy while osteocytes exposed to high doses of GCs or for a prolonged period of time undergo apoptosis.
Collapse
Affiliation(s)
- Wei Yao
- Department of Medicine, University of California at Davis Medical Center Sacramento, CA 95818, USA
| | | | | | | |
Collapse
|
48
|
Viapiana O, Fracassi E, Troplini S, Idolazzi L, Rossini M, Adami S, Gatti D. Sclerostin and DKK1 in primary hyperparathyroidism. Calcif Tissue Int 2013; 92:324-9. [PMID: 23430197 DOI: 10.1007/s00223-012-9665-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 10/18/2012] [Indexed: 12/17/2022]
Abstract
Bone formation is influenced by the Wnt pathway through effects on osteoblast functionality, and these actions are opposed by two antagonists: sclerostin and Dickkopf-1 (DKK1). Decreased levels of serum sclerostin were found after treatment with the PTH analogue teriparatide and in patients with primary hyperparathyroidism (PHPT), while treatment with teriparatide of postmenopausal osteoporosis is associated with increases in serum DKK1. We studied mineral metabolism and Wnt pathway in 21 postmenopausal women affected by PHPT and in 42 age-matched healthy women. Mean serum calcium and PTH were significantly higher and serum phosphates significantly lower in the PHPT group compared with the control group. Serum 25-OH-vitamin D (25OHD) was lower in PHPT patients and 1,25 dihydroxy-vitamin D [1,25(OH)2D] was significantly higher. Patients with PHPT had significantly higher levels of bone alkaline phosphatase (BAP) and of serum C-terminal telopeptides of type I collagene (sCTX). Serum sclerostin in PHPT was significantly lower (-26 %) and serum DKK1 significantly higher (+57 %) than in healthy control subjects. Serum PTH was positively correlated with 1,25OH2D (p < 0.001), BAP (p = 0.036), sCTX (p = 0.003), and DKK1 (p = 0.007) and negatively with 25OHD (p = 0.002) and sclerostin (p = 0.02). In PHPT patients, serum sclerostin was negatively correlated with BAP (p = 0.038) and sCTX (p = 0.07). Patients with PHPT have significantly lower sclerostin and higher DKK1 levels compared with healthy postmenopausal control subjects. Further studies are warranted in order to verify whether the balance between these two opposite effects on Wnt function might help explain the variable bone involvement among patients with PHPT.
Collapse
Affiliation(s)
- Ombretta Viapiana
- Unit of Rheumatology, University of Verona, P.le Scuro 10, 37134, Verona, Italy.
| | | | | | | | | | | | | |
Collapse
|
49
|
van Lierop AH, Hamdy NAT, van Egmond ME, Bakker E, Dikkers FG, Papapoulos SE. Van Buchem disease: clinical, biochemical, and densitometric features of patients and disease carriers. J Bone Miner Res 2013; 28:848-54. [PMID: 23074140 DOI: 10.1002/jbmr.1794] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 09/18/2012] [Accepted: 10/01/2012] [Indexed: 01/08/2023]
Abstract
Van Buchem disease (VBD) is a rare bone sclerosing dysplasia caused by the lack of a regulatory element of the SOST gene, which encodes for sclerostin, an osteocyte-derived negative regulator of bone formation. We studied the demographic, clinical, biochemical, and densitometric features of 15 patients with VBD (12 adults and 3 children) and 28 related carriers of the gene mutation. The most common clinical findings in patients were facial palsy (100%) and various degrees of hearing impairment (93%); raised intracranial pressure had been documented in 20%. The clinical course of the disease appeared to stabilize in adulthood, with the majority of patients reporting no progression of symptoms or development of complications with time. Carriers of the disease had none of the clinical features or complications of the disease. Sclerostin could be detected in the serum in all but 1 VBD patients (mean 8.0 pg/mL; 95% confidence interval [CI], 4.9-11.0 pg/mL), and were lower than those of carriers (mean 28.7 pg/mL; 95% CI, 24.5-32.9 pg/mL; p < 0.001) and healthy controls (mean 40.0 pg/mL; 95% CI, 34.5-41.0 pg/mL; p < 0.). Serum procollagen type 1 amino-terminal propeptide (P1NP) levels were also significantly higher in adult patients (mean 96.0; 95% CI, 54.6-137.4 ng/mL versus mean 47.8; 95% CI, 39.4-56.2 ng/mL, p = 0.003 in carriers and mean 37.8; 95% CI, 34.5-41.0 ng/mL, p = 0.028 in healthy controls) and declined with age. Bone mineral density (BMD) was markedly increased in all patients (mean Z-score 8.7 ± 2.1 and 9.5 ± 1.9 at the femoral neck and spine, respectively); BMD of carriers was significantly lower than that of patients but varied widely (mean Z-scores 0.9 ± 1.0 and 1.3 ± 1.5 at the femoral neck and spine, respectively). Serum sclerostin levels were inversely correlated with serum P1NP levels (r = -0.39, p = 0.018) and BMD values (femoral neck r = -0.69, p < 0.001; lumbar spine r = -0.78, p < 0.001). Our results show that there is a gene-dose effect of the VBD deletion on circulating sclerostin and provide further in vivo evidence of the role of sclerostin in bone formation in humans. The small amounts of sclerostin produced by patients with VBD may explain their milder phenotype compared to that of patients with sclerosteosis, in whom serum sclerostin is undetectable.
Collapse
Affiliation(s)
- Antoon H van Lierop
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
50
|
WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat Med 2013; 19:179-92. [PMID: 23389618 DOI: 10.1038/nm.3074] [Citation(s) in RCA: 1520] [Impact Index Per Article: 126.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 12/18/2012] [Indexed: 12/11/2022]
Abstract
Low bone mass and strength lead to fragility fractures, for example, in elderly individuals affected by osteoporosis or children with osteogenesis imperfecta. A decade ago, rare human mutations affecting bone negatively (osteoporosis-pseudoglioma syndrome) or positively (high-bone mass phenotype, sclerosteosis and Van Buchem disease) have been identified and found to all reside in components of the canonical WNT signaling machinery. Mouse genetics confirmed the importance of canonical Wnt signaling in the regulation of bone homeostasis, with activation of the pathway leading to increased, and inhibition leading to decreased, bone mass and strength. The importance of WNT signaling for bone has also been highlighted since then in the general population in numerous genome-wide association studies. The pathway is now the target for therapeutic intervention to restore bone strength in millions of patients at risk for fracture. This paper reviews our current understanding of the mechanisms by which WNT signalng regulates bone homeostasis.
Collapse
|