1
|
Takegahara N, Kim H, Choi Y. Unraveling the intricacies of osteoclast differentiation and maturation: insight into novel therapeutic strategies for bone-destructive diseases. Exp Mol Med 2024; 56:264-272. [PMID: 38297158 PMCID: PMC10907717 DOI: 10.1038/s12276-024-01157-7] [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: 08/18/2023] [Revised: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 02/02/2024] Open
Abstract
Osteoclasts are the principal cells that efficiently resorb bone. Numerous studies have attempted to reveal the molecular pathways leading to the differentiation and activation of osteoclasts to improve the treatment and prevention of osteoporosis and other bone-destructive diseases. While the cumulative knowledge of osteoclast regulatory molecules, such as receptor activator of nuclear factor-kB ligand (RANKL) and nuclear factor of activated T cells 1 (NFATc1), contributes to the understanding of the developmental progression of osteoclasts, little is known about how the discrete steps of osteoclastogenesis modify osteoclast status but not the absolute number of osteoclasts. The regulatory mechanisms involved in osteoclast maturation but not those involved in differentiation deserve special attention due to their potential use in establishing a more effective treatment strategy: targeting late-phase differentiation while preserving coupled bone formation. Recent studies have shed light on the molecules that govern late-phase osteoclast differentiation and maturation, as well as the metabolic changes needed to adapt to shifting metabolic demands. This review outlines the current understanding of the regulation of osteoclast differentiation, as well as osteoclast metabolic adaptation as a differentiation control mechanism. Additionally, this review introduces molecules that regulate the late-phase osteoclast differentiation and thus minimally impact coupled bone formation.
Collapse
Affiliation(s)
- Noriko Takegahara
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Hyunsoo Kim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
2
|
Everts V, Jansen IDC, de Vries TJ. Mechanisms of bone resorption. Bone 2022; 163:116499. [PMID: 35872106 DOI: 10.1016/j.bone.2022.116499] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 01/08/2023]
Affiliation(s)
- Vincent Everts
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University, Amsterdam, the Netherlands; Department of Anatomy, Dental Faculty, Chulalongkorn University, Bangkok, Thailand.
| | - Ineke D C Jansen
- Department of Periodontology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University, Amsterdam, the Netherlands
| | - Teun J de Vries
- Department of Periodontology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University, Amsterdam, the Netherlands
| |
Collapse
|
3
|
Bolamperti S, Villa I, Rubinacci A. Bone remodeling: an operational process ensuring survival and bone mechanical competence. Bone Res 2022; 10:48. [PMID: 35851054 PMCID: PMC9293977 DOI: 10.1038/s41413-022-00219-8] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 05/02/2022] [Accepted: 05/15/2022] [Indexed: 12/12/2022] Open
Abstract
Bone remodeling replaces old and damaged bone with new bone through a sequence of cellular events occurring on the same surface without any change in bone shape. It was initially thought that the basic multicellular unit (BMU) responsible for bone remodeling consists of osteoclasts and osteoblasts functioning through a hierarchical sequence of events organized into distinct stages. However, recent discoveries have indicated that all bone cells participate in BMU formation by interacting both simultaneously and at different differentiation stages with their progenitors, other cells, and bone matrix constituents. Therefore, bone remodeling is currently considered a physiological outcome of continuous cellular operational processes optimized to confer a survival advantage. Bone remodeling defines the primary activities that BMUs need to perform to renew successfully bone structural units. Hence, this review summarizes the current understanding of bone remodeling and future research directions with the aim of providing a clinically relevant biological background with which to identify targets for therapeutic strategies in osteoporosis.
Collapse
Affiliation(s)
- Simona Bolamperti
- Osteoporosis and Bone and Mineral Metabolism Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milano, Italy
| | - Isabella Villa
- Osteoporosis and Bone and Mineral Metabolism Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milano, Italy
| | - Alessandro Rubinacci
- Osteoporosis and Bone and Mineral Metabolism Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milano, Italy.
| |
Collapse
|
4
|
The Role of the Lysosomal Cl−/H+ Antiporter ClC-7 in Osteopetrosis and Neurodegeneration. Cells 2022; 11:cells11030366. [PMID: 35159175 PMCID: PMC8833911 DOI: 10.3390/cells11030366] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/16/2022] [Accepted: 01/19/2022] [Indexed: 12/04/2022] Open
Abstract
CLC proteins comprise Cl− channels and anion/H+ antiporters involved in several fundamental physiological processes. ClC-7 is a lysosomal Cl−/H+ antiporter that together with its beta subunit Ostm1 has a critical role in the ionic homeostasis of lysosomes and of the osteoclasts’ resorption lacuna, although the specific underlying mechanism has so far remained elusive. Mutations in ClC-7 cause osteopetrosis, but also a form of lysosomal storage disease and neurodegeneration. Interestingly, both loss-of- and gain-of-function mutations of ClC-7 can be pathogenic, but the mechanistic implications of this finding are still unclear. This review will focus on the recent advances in our understanding of the biophysical properties of ClC-7 and of its role in human diseases with a focus on osteopetrosis and neurodegeneration.
Collapse
|
5
|
Rössler U, Hennig AF, Stelzer N, Bose S, Kopp J, Søe K, Cyganek L, Zifarelli G, Ali S, von der Hagen M, Strässler ET, Hahn G, Pusch M, Stauber T, Izsvák Z, Gossen M, Stachelscheid H, Kornak U. Efficient generation of osteoclasts from human induced pluripotent stem cells and functional investigations of lethal CLCN7-related osteopetrosis. J Bone Miner Res 2021; 36:1621-1635. [PMID: 33905594 DOI: 10.1002/jbmr.4322] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/26/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) hold great potential for modeling human diseases and the development of innovative therapeutic approaches. Here, we report on a novel, simplified differentiation method for forming functional osteoclasts from hiPSCs. The three-step protocol starts with embryoid body formation, followed by hematopoietic specification, and finally osteoclast differentiation. We observed continuous production of monocyte-like cells over a period of up to 9 weeks, generating sufficient material for several osteoclast differentiations. The analysis of stage-specific gene and surface marker expression proved mesodermal priming, the presence of monocyte-like cells, and of terminally differentiated multinucleated osteoclasts, able to form resorption pits and trenches on bone and dentine in vitro. In comparison to peripheral blood mononuclear cell (PBMC)-derived osteoclasts hiPSC-derived osteoclasts were larger and contained a higher number of nuclei. Detailed functional studies on the resorption behavior of hiPSC-osteoclasts indicated a trend towards forming more trenches than pits and an increase in pseudoresorption. We used hiPSCs from an autosomal recessive osteopetrosis (ARO) patient (BIHi002-A, ARO hiPSCs) with compound heterozygous missense mutations p.(G292E) and p.(R403Q) in CLCN7, coding for the Cl- /H+ -exchanger ClC-7, for functional investigations. The patient's leading clinical feature was a brain malformation due to defective neuronal migration. Mutant ClC-7 displayed residual expression and retained lysosomal co-localization with OSTM1, the gene coding for the osteopetrosis-associated transmembrane protein 1, but only ClC-7 harboring the mutation p.(R403Q) gave strongly reduced ion currents. An increased autophagic flux in spite of unchanged lysosomal pH was evident in undifferentiated ARO hiPSCs. ARO hiPSC-derived osteoclasts showed an increased size compared to hiPSCs of healthy donors. They were not able to resorb bone, underlining a loss-of-function effect of the mutations. In summary, we developed a highly reproducible, straightforward hiPSC-osteoclast differentiation protocol. We demonstrated that osteoclasts differentiated from ARO hiPSCs can be used as a disease model for ARO and potentially also other osteoclast-related diseases. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Uta Rössler
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Anna Floriane Hennig
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Freie Universität Berlin, Berlin, Germany.,Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Nina Stelzer
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Shroddha Bose
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Johannes Kopp
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Freie Universität Berlin, Berlin, Germany.,Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense C, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense M, Denmark.,Department of Molecular Medicine, University of Southern Denmark, Odense M, Denmark
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | | | - Salaheddine Ali
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Maja von der Hagen
- Abteilung Neuropädiatrie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Elisabeth Tamara Strässler
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Gabriele Hahn
- Institut und Poliklinik für Radiologische Diagnostik, Medizinische Fakultät Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
| | | | - Tobias Stauber
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Department of Human Medicine, and Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Zsuzsanna Izsvák
- Max-Delbrück-Center for Molecular Medicine (MDC), Helmholtz Association, Berlin, Germany
| | - Manfred Gossen
- Berlin-Brandenburg Center for Regenerative Therapies, Charité Virchow Campus, Berlin, Germany.,Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Harald Stachelscheid
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Berlin Institute of Health (BIH), BIH Stem Cell Core Facility, Berlin, Germany
| | - Uwe Kornak
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
6
|
Influences of the IL-6 cytokine family on bone structure and function. Cytokine 2021; 146:155655. [PMID: 34332274 DOI: 10.1016/j.cyto.2021.155655] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 01/12/2023]
Abstract
The IL-6 family of cytokines comprises a large group of cytokines that all act via the formation of a signaling complex that includes the glycoprotein 130 (gp130) receptor. Despite this, many of these cytokines have unique roles that regulate the activity of bone forming osteoblasts, bone resorbing osteoclasts, bone-resident osteocytes, and cartilage cells (chondrocytes). These include specific functions in craniofacial development, longitudinal bone growth, and the maintenance of trabecular and cortical bone structure, and have been implicated in musculoskeletal pathologies such as craniosynostosis, osteoporosis, rheumatoid arthritis, osteoarthritis, and heterotopic ossifications. This review will work systematically through each member of this family and provide an overview and an update on the expression patterns and functions of each of these cytokines in the skeleton, as well as their negative feedback pathways, particularly suppressor of cytokine signaling 3 (SOCS3). The specific cytokines described are interleukin 6 (IL-6), interleukin 11 (IL-11), oncostatin M (OSM), leukemia inhibitory factor (LIF), cardiotrophin 1 (CT-1), ciliary neurotrophic factor (CNTF), cardiotrophin-like cytokine factor 1 (CLCF1), neuropoietin, humanin and interleukin 27 (IL-27).
Collapse
|
7
|
Canalis E, Zanotti S, Schilling L, Eller T, Yu J. Activation of Notch3 in osteoblasts/osteocytes causes compartment-specific changes in bone remodeling. J Biol Chem 2021; 296:100583. [PMID: 33774049 PMCID: PMC8086145 DOI: 10.1016/j.jbc.2021.100583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/12/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
Notch receptors maintain skeletal homeostasis. NOTCH1 and 2 have been studied for their effects on bone remodeling. Although NOTCH3 plays a significant role in vascular physiology, knowledge about its function in other cellular environments, including bone, is limited. The present study was conducted to establish the function of NOTCH3 in skeletal cells using models of Notch3 misexpression. Microcomputed tomography demonstrated that Notch3 null mice did not have appreciable bone phenotypes. To study the effects of the NOTCH3 activation in the osteoblast lineage, BGLAP-Cre or Dmp1-Cre transgenics were crossed with RosaNotch3 mice, where the NOTCH3 intracellular domain is expressed following the removal of a loxP-flanked STOP cassette. Microcomputed tomography demonstrated that BGLAP-Cre;RosaNotch3 and Dmp1-Cre;RosaNotch3 mice of both sexes exhibited an increase in trabecular bone and in connectivity, with a decrease in cortical bone and increased cortical porosity. Histological analysis revealed a decrease in osteoclast number and bone resorption in trabecular bone and an increase in osteoclast number and void or pore area in cortical bone of RosaNotch3 mice. Bone formation was either decreased or could not be determined in Cre;RosaNotch3 mice. NOTCH3 activation in osteoblasts inhibited Alpl (alkaline phosphatase) and Bglap (osteocalcin) and induced Tnfsf11 (RANKL) and Tnfrsf11b (osteoprotegerin) mRNA, possibly explaining the trabecular bone phenotype. However, NOTCH3 induced Tnfsf11 and suppressed Tnfrsf11b in osteocytes, possibly explaining the cortical porosity. In conclusion, basal NOTCH3 is dispensable for skeletal homeostasis, whereas activation of NOTCH3 in osteoblasts/osteocytes inhibits osteoclastogenesis and bone resorption in cancellous bone but increases intracortical remodeling and causes cortical porosity.
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, USA; Department of Medicine, UConn Health, Farmington, Connecticut, USA; UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, USA.
| | - Stefano Zanotti
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, USA; UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, USA
| | - Lauren Schilling
- UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, USA
| | - Tabitha Eller
- UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, USA
| | - Jungeun Yu
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, USA; UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
8
|
Vacher J, Bruccoleri M, Pata M. Ostm1 from Mouse to Human: Insights into Osteoclast Maturation. Int J Mol Sci 2020; 21:ijms21165600. [PMID: 32764302 PMCID: PMC7460669 DOI: 10.3390/ijms21165600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
The maintenance of bone mass is a dynamic process that requires a strict balance between bone formation and resorption. Bone formation is controlled by osteoblasts, while osteoclasts are responsible for resorption of the bone matrix. The opposite functions of these cell types have to be tightly regulated not only during normal bone development, but also during adult life, to maintain serum calcium homeostasis and sustain bone integrity to prevent bone fractures. Disruption of the control of bone synthesis or resorption can lead to an over accumulation of bone tissue in osteopetrosis or conversely to a net depletion of the bone mass in osteoporosis. Moreover, high levels of bone resorption with focal bone formation can cause Paget’s disease. Here, we summarize the steps toward isolation and characterization of the osteopetrosis associated trans-membrane protein 1 (Ostm1) gene and protein, essential for proper osteoclast maturation, and responsible when mutated for the most severe form of osteopetrosis in mice and humans.
Collapse
Affiliation(s)
- Jean Vacher
- Institut de Recherches Cliniques de Montreal (IRCM), Montreal, QC H2W 1R7, Canada; (M.B.); (M.P.)
- Departement de Medecine, Universite de Montreal, Montreal, QC H2W 1R7, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC H3A 1A3, Canada
- Correspondence:
| | - Michael Bruccoleri
- Institut de Recherches Cliniques de Montreal (IRCM), Montreal, QC H2W 1R7, Canada; (M.B.); (M.P.)
- Departement de Medecine, Universite de Montreal, Montreal, QC H2W 1R7, Canada
| | - Monica Pata
- Institut de Recherches Cliniques de Montreal (IRCM), Montreal, QC H2W 1R7, Canada; (M.B.); (M.P.)
| |
Collapse
|
9
|
Deng Z, Li W, Xu J, Yu M, Li D, Tan Q, Wang D, Chen L, Wang L. ClC-3 chloride channels are involved in estradiol regulation of bone formation by MC3T3-E1 osteoblasts. J Cell Biochem 2019; 120:8366-8375. [PMID: 30506861 DOI: 10.1002/jcb.28121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/31/2018] [Indexed: 01/24/2023]
Abstract
Evidence has been reported by us and others supporting the important roles of chloride channels in a number of osteoblast cell functions. The ClC-3 chloride channel is activated by estradiol binding to estrogen receptor alpha on the cell membranes of osteoblasts. However, the functions of these chloride channels in estrogen regulation of osteoblast metabolism remain unclear. In the present study, the roles of chloride channels in estrogen regulation of osteoblasts were investigated in the osteoblastic cell line MC3T3-E1. Estrogen 17β-estradiol enhanced collagen I protein expression, alkaline phosphatase activity, and mineralization were inhibited, by chloride channel blockers. Estradiol promoted ClC-3 chloride channel protein expression. Silencing of ClC-3 chloride channel expression prevented the elevation of osteodifferentiation in osteoblasts, which were regulated by estrogen. These data suggest that estrogen can regulate bone formation by activating ClC-3 chloride channels and the activation of ClC-3 chloride channels can enhance the osteodifferentiation in osteoblasts.
Collapse
Affiliation(s)
- Zhiqin Deng
- Hand and Foot Surgery Department, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, China.,Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Wencui Li
- Hand and Foot Surgery Department, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, China
| | - Jianying Xu
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Meishen Yu
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Duan Li
- Hand and Foot Surgery Department, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, China
| | - Qiuchan Tan
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Daping Wang
- Hand and Foot Surgery Department, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, China
| | - Lixin Chen
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Liwei Wang
- Department of Physiology, Medical College, Jinan University, Guangzhou, China.,International School, Jinan University, Guangzhou, China
| |
Collapse
|
10
|
Butscheidt S, Rolvien T, Kornak U, Schmidt FN, Schinke T, Amling M, Oheim R. Clinical Significance of DXA and HR-pQCT in Autosomal Dominant Osteopetrosis (ADO II). Calcif Tissue Int 2018; 102:41-52. [PMID: 29018903 DOI: 10.1007/s00223-017-0332-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/21/2017] [Indexed: 12/29/2022]
Abstract
The main hallmark of high bone mass (HBM) disorders is increased bone mineral density, potentially visible in conventional radiographs and quantifiable by other radiographic methods. While one of the most common forms of HBM is CLCN7-related autosomal dominant osteopetrosis type II (ADO II), there is no consensus on diagnostic thresholds. We therefore wanted to assess whether CLCN7-osteopetrosis patients differ from benign HBM cases in terms of (1) bone mineral density, (2) bone structure, and (3) microarchitectural abnormalities. 16 patients meeting the criteria of HBM (DXA T/Z-score ≥ 2.5 at all sites) were included in this retrospective study. Osteologic assessment using dual-energy X-ray absorptiometry (DXA), high-resolution peripheral quantitative computed tomography (HR-pQCT), and serum analyses was performed. The presence of CLCN7 and/or other HBM gene mutations affecting bone mass were tested using a custom designed bone panel. While a DXA threshold for ADO II could be implemented (DXA Z-score ≥ + 6.0), the differences in bone microarchitecture were of lesser extent compared to the benign HBM group. All adult patients with ADO II suffered from elevated fracture rates independent from Z-score. In HR-pQCT, structural alterations, such as bone islets were found only inconsistently. In cases of HBM, a DXA Z-score ≥ 6 may be indicative for an inheritable HBM disorder, such as ADO II. Microarchitectural bone alterations might represent local microfracture repair or accumulation of cartilage remnants due to impaired osteoclast function, but seem not to be correlated with fracture risk.
Collapse
Affiliation(s)
- Sebastian Butscheidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Uwe Kornak
- Institute of Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité Universitätsmedizin Berlin, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Felix N Schmidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany.
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| |
Collapse
|
11
|
Li BY, Gao YH, Pei JR, Yang YM, Zhang W, Sun DJ. ClC-7/Ostm1 contribute to the ability of tea polyphenols to maintain bone homeostasis in C57BL/6 mice, protecting against fluorosis. Int J Mol Med 2017; 39:1155-1163. [PMID: 28339032 PMCID: PMC5403613 DOI: 10.3892/ijmm.2017.2933] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 02/21/2017] [Indexed: 01/09/2023] Open
Abstract
Epidemiological investigations indicate that certain ingredients in tea bricks can antagonize the adverse effects of fluoride. Tea polyphenols (TPs), the most bioactive ingredient in tea bricks, have been demonstrated to be potent bone-supporting agents. ClC‑7 is known to be crucial for osteoclast (OC) bone resorption. Thus, in this study, we investigated the potential protective effects of TPs against fluorosis using a mouse model and explored the underlying mechanisms with particular focus on ClC‑7. A total of 40, healthy, 3‑week‑old male C57BL/6 mice were randomly divided into 4 groups (n=10/group) by weight as follows: distilled water (control group), 100 mg/l fluoridated water (F group), water containing 10 g/l TPs (TP group) and water containing 100 mg/l fluoride and 10 g/l TPs (F + TP group). After 15 weeks, and after the mice were sacrificed, the long bones were removed and bone marrow-derived macrophages were cultured ex vivo in order to perform several experiments. OCs were identified and counted by tartrate‑resistant acid phosphatase (TRAP) staining. The consumption of fluoride resulted in severe fluorosis and in an impaired OC function [impaired bone resorption, and a low mRNA expression of nuclear factor of activated T-cells 1 (NFATc1), ATPase H+ transporting V0 subunit D2 (ATP6v0d2) and osteopetrosis‑associated transmembrane protein 1 (Ostm1)]. In the F + TP group, fluorosis was attenuated and OC function was restored, but not the high bone fluoride content. Compared with the F group, mature OCs in the F + TP group expressed higher mRNA levels of ClC‑7 and Ostm1; the transportation and retaining of Cl‑ was improved, as shown by the fluorescence intensity experiment. On the whole, our findings indicate that TPs mitigate fluorosis in C57BL/6 mice by regulating OC bone resorption. Fluoride inhibits OC resorption by inhibiting ClC‑7 and Ostm1, whereas TPs attenuate this inhibitory effect of fluoride.
Collapse
Affiliation(s)
- Bing-Yun Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yan-Hui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jun-Rui Pei
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yan-Mei Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Wei Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Dian-Jun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
12
|
Alam I, McQueen AK, Acton D, Reilly AM, Gerard-O'Riley RL, Oakes DK, Kasipathi C, Huffer A, Wright WB, Econs MJ. Phenotypic severity of autosomal dominant osteopetrosis type II (ADO2) mice on different genetic backgrounds recapitulates the features of human disease. Bone 2017; 94:34-41. [PMID: 27746321 DOI: 10.1016/j.bone.2016.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 10/11/2016] [Accepted: 10/13/2016] [Indexed: 10/20/2022]
Abstract
Autosomal dominant osteopetrosis type II (ADO2) is a heritable osteosclerotic bone disorder due to dysfunctional osteoclast activity. ADO2 is caused by missense mutations in the chloride channel 7 (CLCN7) gene characterized by osteosclerosis with multiple fractures. ADO2 can result in osteomyelitis, visual loss and bone marrow failure. Currently, there is no cure for ADO2, and until recently no appropriate animal model of ADO2 existed to understand better the pathogenesis of this disease and to test new therapies. Therefore, we created ADO2 knock-in mouse model with a G213R (human homolog of G215R) missense mutation in the Clcn7 gene on 129S1 background, and demonstrated that this mouse model phenocopies human ADO2. As ADO2 gives rise to incomplete penetrance (66%) in human and marked phenotypic variability is observed among patients with the same mutation, we hypothesized that the severity and penetrance of ADO2 will also vary in mouse models on different genetic backgrounds. To test this, we created ADO2 mouse models in DBA/D2, C57BL/6J/B6 and Balb/c strains, and compared bone phenotypes and performed serum biochemical analysis between strain- and age-matched wild-type (WT) and ADO2 mice. At 3months of age, whole body aBMD was higher (4-7% in male; 1-5% in female) in the ADO2 mice compared to their wild-type littermates. In addition, ADO2 male mice on 129 background displayed highest percent increase of BV/TV (106%), followed by D2 (92%), B6 (46%), and Balb/c (33%) compared to strain-matched wild-type mice. We observed similar differences for BV/TV between ADO2 and wild-type mice on different genetic backgrounds in female: 129 (96%)>D2 (73%)>Balb/c (39%) and B6 (36%). Serum calcium, phosphorus, alkaline phosphatase and P1NP levels were similar in the WT and ADO2 mice on all genetic backgrounds but TRAP was higher (76% to 220% in male; 33-95% in female) and CTX/TRAP ratio was lower (39-65% in male and 3-41% in female) in the ADO2 mice compared to their strain-matched wild-type littermates. We also found that young (3months) ADO2 mice on 129S1 background exhibited 200% higher trabecular BV/TV whereas old (18months) ADO2 mice displayed 400-700% higher BV/TV compared to their age-matched wild-type controls. In summary, phenotypic severity in ADO2 mice varied markedly on different genetic backgrounds (129>D2>Balb/c>B6) and became more pronounced with age, which resembles the wide variations in phenotype observed in ADO2 patients. These mouse models will help us to identify genes/factors that influence severity and penetrance of ADO2, and test innovative therapies to treat this disease.
Collapse
Affiliation(s)
- Imranul Alam
- Medicine, Indiana University School of Medicine, IN, USA.
| | - Amie K McQueen
- Medicine, Indiana University School of Medicine, IN, USA
| | - Dena Acton
- Medicine, Indiana University School of Medicine, IN, USA
| | | | | | - Dana K Oakes
- Medicine, Indiana University School of Medicine, IN, USA
| | | | - Abigail Huffer
- Medicine, Indiana University School of Medicine, IN, USA
| | | | - Michael J Econs
- Medicine, Indiana University School of Medicine, IN, USA; Medical and Molecular Genetics, Indiana University School of Medicine, IN, USA
| |
Collapse
|
13
|
Fujiwara T, Ye S, Castro-Gomes T, Winchell CG, Andrews NW, Voth DE, Varughese KI, Mackintosh SG, Feng Y, Pavlos N, Nakamura T, Manolagas SC, Zhao H. PLEKHM1/DEF8/RAB7 complex regulates lysosome positioning and bone homeostasis. JCI Insight 2016; 1:e86330. [PMID: 27777970 DOI: 10.1172/jci.insight.86330] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Mutations of the Plekhm1 gene in humans and rats cause osteopetrosis, an inherited bone disease characterized by diminished bone resorption by osteoclasts. PLEKHM1 binds to RAB7 and is critical for lysosome trafficking. However, the molecular mechanisms by which PLEKHM1 regulates lysosomal pathways remain unknown. Here, we generated germline and conditional Plekhm1-deficient mice. These mice displayed no overt abnormalities in major organs, except for an increase in trabecular bone mass. Furthermore, loss of PLEKHM1 abrogated the peripheral distribution of lysosomes and bone resorption in osteoclasts. Mechanistically, we indicated that DEF8 interacts with PLEKHM1 and promotes its binding to RAB7, whereas the binding of FAM98A and NDEL1 with PLEKHM1 connects lysosomes to microtubules. Importantly, suppression of these proteins results in lysosome positioning and bone resorption defects similar to those of Plekhm1-null osteoclasts. Thus, PLHKEM1, DEF8, FAM98A, and NDEL1 constitute a molecular complex that regulates lysosome positioning and secretion through RAB7.
Collapse
Affiliation(s)
- Toshifumi Fujiwara
- Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Shiqiao Ye
- Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Thiago Castro-Gomes
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | | | - Norma W Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | | | | | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Yunfeng Feng
- Department of Pathology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Nathan Pavlos
- Center for Orthopedic Research, Dentistry and Health Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Takashi Nakamura
- Department of Biochemistry & Integrative Medical Biology, School of Medicine, Keio University, Tokyo, Japan
| | - Stavros C Manolagas
- Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Haibo Zhao
- Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Physiology and Biophysics, and
| |
Collapse
|
14
|
Brömme D, Panwar P, Turan S. Cathepsin K osteoporosis trials, pycnodysostosis and mouse deficiency models: Commonalities and differences. Expert Opin Drug Discov 2016; 11:457-72. [DOI: 10.1517/17460441.2016.1160884] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Dieter Brömme
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, Canada
| | - Preety Panwar
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, Canada
| | - Serap Turan
- Department of Pediatric Endocrinology, Marmara University, Istanbul, Turkey
| |
Collapse
|
15
|
ClC-7 Deficiency Impairs Tooth Development and Eruption. Sci Rep 2016; 6:19971. [PMID: 26829236 PMCID: PMC4734291 DOI: 10.1038/srep19971] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 12/22/2015] [Indexed: 12/21/2022] Open
Abstract
CLCN7 gene encodes the voltage gated chloride channel 7 (ClC-7) in humans. The mutations in CLCN7 have been associated with osteopetrosis in connection to the abnormal osteoclasts functions. Previously, we found that some osteopetrosis patients with CLCN7 mutations suffered from impacted teeth and root dysplasia. Here we set up two in vivo models under a normal or an osteoclast-poor environment to investigate how ClC-7 affects tooth development and tooth eruption. Firstly, chitosan-Clcn7-siRNA nanoparticles were injected around the first maxillary molar germ of newborn mice and caused the delay of tooth eruption and deformed tooth with root dysplasia. Secondly, E13.5 molar germs infected with Clcn7 shRNA lentivirus were transplanted under the kidney capsule and presented the abnormal changes in dentin structure, periodontal tissue and cementum. All these teeth changes have been reported in the patients with CLCN7 mutation. In vitro studies of ameloblasts, odontoblasts and dental follicle cells (DFCs) were conducted to explore the involved mechanism. We found that Clcn7 deficiency affect the differentiation of these cells, as well as the interaction between DFCs and osteoclasts through RANKL/OPG pathway. We conclude that ClC-7 may affect tooth development by directly targeting tooth cells, and regulate tooth eruption through DFC mediated osteoclast pathway.
Collapse
|
16
|
Tonna S, Poulton IJ, Taykar F, Ho PWM, Tonkin B, Crimeen-Irwin B, Tatarczuch L, McGregor NE, Mackie EJ, Martin TJ, Sims NA. Chondrocytic ephrin B2 promotes cartilage destruction by osteoclasts in endochondral ossification. Development 2016; 143:648-57. [PMID: 26755702 DOI: 10.1242/dev.125625] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 12/24/2015] [Indexed: 12/17/2022]
Abstract
The majority of the skeleton arises by endochondral ossification, whereby cartilaginous templates expand and are resorbed by osteoclasts then replaced by osteoblastic bone formation. Ephrin B2 is a receptor tyrosine kinase expressed by osteoblasts and growth plate chondrocytes that promotes osteoblast differentiation and inhibits osteoclast formation. We investigated the role of ephrin B2 in endochondral ossification using Osx1Cre-targeted gene deletion. Neonatal Osx1Cre.Efnb2(Δ/Δ) mice exhibited a transient osteopetrosis demonstrated by increased trabecular bone volume with a high content of growth plate cartilage remnants and increased cortical thickness, but normal osteoclast numbers within the primary spongiosa. Osteoclasts at the growth plate had an abnormal morphology and expressed low levels of tartrate-resistant acid phosphatase; this was not observed in more mature bone. Electron microscopy revealed a lack of sealing zones and poor attachment of Osx1Cre.Efnb2(Δ/Δ) osteoclasts to growth plate cartilage. Osteoblasts at the growth plate were also poorly attached and impaired in their ability to deposit osteoid. By 6 months of age, trabecular bone mass, osteoclast morphology and osteoid deposition by Osx1Cre.Efnb2(Δ/Δ) osteoblasts were normal. Cultured chondrocytes from Osx1Cre.Efnb2(Δ/Δ) neonates showed impaired support of osteoclastogenesis but no significant change in Rankl (Tnfsf11) levels, whereas Adamts4 levels were significantly reduced. A population of ADAMTS4(+) early hypertrophic chondrocytes seen in controls was absent from Osx1Cre.Efnb2(Δ/Δ) neonates. This suggests that Osx1Cre-expressing cells, including hypertrophic chondrocytes, are dependent on ephrin B2 for their production of cartilage-degrading enzymes, including ADAMTS4, and this might be required for attachment of osteoclasts and osteoblasts to the cartilage surface during endochondral ossification.
Collapse
Affiliation(s)
- Stephen Tonna
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria 3065, Australia
| | - Ingrid J Poulton
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Farzin Taykar
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Patricia W M Ho
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Brett Tonkin
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | | | - Liliana Tatarczuch
- The University of Melbourne, Faculty of Veterinary and Agricultural Sciences, Parkville 3010, Australia
| | - Narelle E McGregor
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Eleanor J Mackie
- The University of Melbourne, Faculty of Veterinary and Agricultural Sciences, Parkville 3010, Australia
| | - T John Martin
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria 3065, Australia
| | - Natalie A Sims
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria 3065, Australia
| |
Collapse
|
17
|
Johnson RW, McGregor NE, Brennan HJ, Crimeen-Irwin B, Poulton IJ, Martin TJ, Sims NA. Glycoprotein130 (Gp130)/interleukin-6 (IL-6) signalling in osteoclasts promotes bone formation in periosteal and trabecular bone. Bone 2015; 81:343-351. [PMID: 26255596 DOI: 10.1016/j.bone.2015.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 07/27/2015] [Accepted: 08/04/2015] [Indexed: 12/31/2022]
Abstract
Interleukin-6 (IL-6) and interleukin-11 (IL-11) receptors (IL-6R and IL-11R, respectively) are both expressed in osteoclasts and transduce signal via the glycoprotein130 (gp130) co-receptor, but the physiological role of this pathway is unclear. To determine the critical roles of gp130 signalling in the osteoclast, we generated mice using cathepsin K Cre (CtskCre) to disrupt gp130 signalling in osteoclasts. Bone marrow macrophages from CtskCre.gp130(f/f) mice generated more osteoclasts in vitro than cells from CtskCre.gp130(w/w) mice; these osteoclasts were also larger and had more nuclei than controls. While no increase in osteoclast numbers was observed in vivo, osteoclasts on trabecular bone surfaces of CtskCre.gp130(f/f) mice were more spread out than in control mice, but had no functional defect detectable by serum CTX1 levels or trabecular bone cartilage remnants. However, trabecular osteoblast number and mineralising surfaces were significantly lower in male CtskCre.gp130(f/f) mice compared to controls, and this was associated with a significantly lower trabecular bone volume at 12 weeks of age. Furthermore, CtskCre.gp130(f/f) mice exhibited greatly suppressed periosteal bone formation at this age, indicated by significant reductions in both double-labelled surface and mineral apposition rate. By 26 weeks of age, CtskCre.gp130(f/f) mice exhibited narrower femora, with lower periosteal and endocortical perimeters than CtskCre.gp130(w/w) controls. Since IL-6 and IL-11R global knockout mice exhibited a similar reduction in femoral width, we also assessed periosteal bone formation in those strains, and found bone forming surfaces were also reduced in male IL-6 null mice. These data suggest that IL-6/gp130 signalling in the osteoclast is not essential for normal bone resorption in vivo, but maintains both trabecular and periosteal bone formation in male mice by promoting osteoblast activity through the stimulation of osteoclast-derived "coupling factors" and "osteotransmitters", respectively.
Collapse
Affiliation(s)
| | | | - Holly J Brennan
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | | | - Ingrid J Poulton
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - T John Martin
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, VIC, Australia
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, VIC, Australia.
| |
Collapse
|
18
|
Henriksen K, Thudium CS, Christiansen C, Karsdal MA. Novel targets for the prevention of osteoporosis - lessons learned from studies of metabolic bone disorders. Expert Opin Ther Targets 2015; 19:1575-84. [PMID: 25960169 DOI: 10.1517/14728222.2015.1045415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Osteoporosis is a major health care problem, and whereas efficacious treatments for vertebral fracture reduction are available for osteoporosis patients, these therapies are still limited with respect to capacity for restoration of bone loss, as well as efficacy on non-vertebral fractures, such as hip fractures, which are the source of morbidity and mortality. AREAS COVERED Studies of rare bone diseases in humans, such as osteopetrosis, sclerosteosis, pycnodysostosis and more, have shed light on a series of drug targets in bone that have the potential to result in therapies for osteoporosis with novel mechanisms of action, and the potential to improve the standard of care substantially. We focus on how they are separated from classic treatments for osteoporosis, in terms of novel modes of action, additional beneficial effects on bone turnover and importantly also safety. We focus on the status of anti-sclerostin antibodies, novel parathyroid hormone-related protein analogs, inhibitors of cathepsin K and ClC-7 in osteoclasts, all of which are currently in development. EXPERT OPINION There is a good possibility that the treatment of osteoporosis will be greatly improved within the coming years; however, with numerous effective and safe drugs already available careful attention to the safety of these novel candidates is crucial.
Collapse
Affiliation(s)
- Kim Henriksen
- a Department of Musculoskeletal Diseases, Nordic Bioscience Biomarkers and Research , Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Christian Schneider Thudium
- a Department of Musculoskeletal Diseases, Nordic Bioscience Biomarkers and Research , Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Claus Christiansen
- a Department of Musculoskeletal Diseases, Nordic Bioscience Biomarkers and Research , Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Morten Asser Karsdal
- a Department of Musculoskeletal Diseases, Nordic Bioscience Biomarkers and Research , Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| |
Collapse
|
19
|
Abdelmagid SM, Sondag GR, Moussa FM, Belcher JY, Yu B, Stinnett H, Novak K, Mbimba T, Khol M, Hankenson KD, Malcuit C, Safadi FF. Mutation in Osteoactivin Promotes Receptor Activator of NFκB Ligand (RANKL)-mediated Osteoclast Differentiation and Survival but Inhibits Osteoclast Function. J Biol Chem 2015; 290:20128-46. [PMID: 25837253 DOI: 10.1074/jbc.m114.624270] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Indexed: 12/29/2022] Open
Abstract
We previously reported on the importance of osteoactivin (OA/Gpnmb) in osteogenesis. In this study, we examined the role of OA in osteoclastogenesis, using mice with a nonsense mutation in the Gpnmb gene (D2J) and wild-type controls (D2J/Gpnmb(+)). In these D2J mice, micro-computed tomography and histomorphometric analyses revealed increased cortical thickness, whereas total porosity and eroded surface were significantly reduced in D2J mice compared with wild-type controls, and these results were corroborated by lower serum levels of CTX-1. Contrary to these observations and counterintuitively, temporal gene expression analyses supported up-regulated osteoclastogenesis in D2J mice and increased osteoclast differentiation rates ex vivo, marked by increased number and size. The finding that MAPK was activated in early differentiating and mature D2J osteoclasts and that survival of D2J osteoclasts was enhanced and mediated by activation of the AKT-GSK3β pathway supports this observation. Furthermore, this was abrogated by the addition of recombinant OA to cultures, which restored osteoclastogenesis to wild-type levels. Moreover, mix and match co-cultures demonstrated an induction of osteoclastogenesis in D2J osteoblasts co-cultured with osteoclasts of D2J or wild-type. Last, in functional osteo-assays, we show that bone resorption activity of D2J osteoclasts is dramatically reduced, and these osteoclasts present an abnormal ruffled border over the bone surface. Collectively, these data support a model whereby OA/Gpnmb acts as a negative regulator of osteoclast differentiation and survival but not function by inhibiting the ERK/AKT signaling pathways.
Collapse
Affiliation(s)
- Samir M Abdelmagid
- From the Department of Anatomy and Neurobiology, Northeast Ohio Medical University College of Medicine, Rootstown, Ohio 44272
| | - Gregory R Sondag
- From the Department of Anatomy and Neurobiology, Northeast Ohio Medical University College of Medicine, Rootstown, Ohio 44272, the School of Biomedical Sciences and
| | - Fouad M Moussa
- From the Department of Anatomy and Neurobiology, Northeast Ohio Medical University College of Medicine, Rootstown, Ohio 44272, the School of Biomedical Sciences and
| | - Joyce Y Belcher
- the Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, and
| | - Bing Yu
- From the Department of Anatomy and Neurobiology, Northeast Ohio Medical University College of Medicine, Rootstown, Ohio 44272
| | - Hilary Stinnett
- From the Department of Anatomy and Neurobiology, Northeast Ohio Medical University College of Medicine, Rootstown, Ohio 44272
| | - Kimberly Novak
- From the Department of Anatomy and Neurobiology, Northeast Ohio Medical University College of Medicine, Rootstown, Ohio 44272
| | - Thomas Mbimba
- From the Department of Anatomy and Neurobiology, Northeast Ohio Medical University College of Medicine, Rootstown, Ohio 44272
| | - Matthew Khol
- From the Department of Anatomy and Neurobiology, Northeast Ohio Medical University College of Medicine, Rootstown, Ohio 44272
| | - Kurt D Hankenson
- the Department of Clinical Studies, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania 19104
| | - Christopher Malcuit
- the School of Biomedical Sciences and Department of Biological Sciences, Kent State University, Kent, Ohio 44240
| | - Fayez F Safadi
- From the Department of Anatomy and Neurobiology, Northeast Ohio Medical University College of Medicine, Rootstown, Ohio 44272, the School of Biomedical Sciences and
| |
Collapse
|
20
|
Wen X, Lacruz RS, Paine ML. Dental and Cranial Pathologies in Mice Lacking the Cl(-) /H(+) -Exchanger ClC-7. Anat Rec (Hoboken) 2015; 298:1502-8. [PMID: 25663454 DOI: 10.1002/ar.23118] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 12/01/2014] [Accepted: 12/05/2014] [Indexed: 11/10/2022]
Abstract
ClC-7 is a 2Cl(-) /1H(+) -exchanger expressed at late endosomes and lysosomes, as well as the ruffled border of osteoclasts. ClC-7 deficiencies in mice and humans lead to impaired osteoclast function and therefore osteopetrosis. Failure of tooth eruption is also apparent in ClC-7 mutant animals, and this has been attributed to the osteoclast dysfunction and the subsequent defect in alveolar bone resorptive activity surrounding tooth roots. Ameloblasts also express ClC-7, and this study aims to determine the significance of ClC-7 in enamel formation by examining the dentitions of ClC-7 mutant mice. Micro-CT analysis revealed that the molar teeth of 3-week old ClC-7 mutant mice had no roots, and the incisors were smaller than their age-matched controls. Despite these notable developmental differences, the enamel and dentin densities of the mutant mice were comparable to those of the wild-type littermates. Scanning electron microscopy showed normal enamel crystallite and prismatic organization in the ClC-7 mutant mice, although the enamel was thinner (hypoplastic) than in controls. These results suggested that ClC-7 was not critical to enamel and dentin formation, and the observed tooth defects may be related more to a resulting alveolar bone phenotype. Micro-CT analysis also revealed abnormal features in the calvarial bones of the mutant mice. The cranial sutures in ClC-7 mutant mice remained open compared to the closed sutures seen in the control mice at 3 weeks. These data demonstrate that ClC-7 deficiency impacts the development of the dentition and calvaria, but does not significantly disrupt amelogenesis.
Collapse
Affiliation(s)
- Xin Wen
- Herman Ostrow School of Dentistry of USC, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, USA
| | - Rodrigo S Lacruz
- Department of Basic Science and Craniofacial Biology, College of Dentistry, New York University, New York, New York, USA
| | - Michael L Paine
- Herman Ostrow School of Dentistry of USC, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, USA
| |
Collapse
|
21
|
Thudium CS, Moscatelli I, Flores C, Thomsen JS, Brüel A, Gudmann NS, Hauge EM, Karsdal MA, Richter J, Henriksen K. A comparison of osteoclast-rich and osteoclast-poor osteopetrosis in adult mice sheds light on the role of the osteoclast in coupling bone resorption and bone formation. Calcif Tissue Int 2014; 95:83-93. [PMID: 24838599 DOI: 10.1007/s00223-014-9865-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 04/28/2014] [Indexed: 01/15/2023]
Abstract
Osteopetrosis due to lack of acid secretion by osteoclasts is characterized by abolished bone resorption, increased osteoclast numbers, but normal or even increased bone formation. In contrast, osteoclast-poor osteopetrosis appears to have less osteoblasts and reduced bone formation, indicating that osteoclasts are important for regulating osteoblast activity. To illuminate the role of the osteoclast in controlling bone remodeling, we transplanted irradiated skeletally mature 3-month old wild-type mice with hematopoietic stem cells (HSCs) to generate either an osteoclast-rich or osteoclast-poor adult osteopetrosis model. We used fetal liver HSCs from (1) oc/oc mice, (2) RANK KO mice, and (3) compared these to wt control cells. TRAP5b activity, a marker of osteoclast number and size, was increased in the oc/oc recipients, while a significant reduction was seen in the RANK KO recipients. In contrast, the bone resorption marker CTX-I was similarly decreased in both groups. Both oc/oc and Rank KO recipients developed a mild osteopetrotic phenotype. However, the osteoclast-rich oc/oc recipients showed higher trabecular bone volume (40 %), increased bone strength (66 %), and increased bone formation rate (54 %) in trabecular bone, while RANK KO recipients showed only minor trends compared to control recipients. We here show that maintaining non-resorbing osteoclasts, as opposed to reducing the osteoclasts, leads to increased bone formation, bone volume, and ultimately higher bone strength in vivo, which indicates that osteoclasts are sources of anabolic molecules for the osteoblasts.
Collapse
|
22
|
Touaitahuata H, Cres G, de Rossi S, Vives V, Blangy A. The mineral dissolution function of osteoclasts is dispensable for hypertrophic cartilage degradation during long bone development and growth. Dev Biol 2014; 393:57-70. [PMID: 24992711 DOI: 10.1016/j.ydbio.2014.06.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 06/13/2014] [Accepted: 06/18/2014] [Indexed: 11/26/2022]
Abstract
During long bone development and post-natal growth, the cartilaginous model of the skeleton is progressively replaced by bone, a process known as endochondral ossification. In the primary spongiosa, osteoclasts degrade the mineralized cartilage produced by hypertrophic chondrocytes to generate cartilage trabeculae that osteoblasts embed in bone matrix. This leads to the formation of the trabecular bone network of the secondary spongiosa that will undergo continuous remodeling. Osteoclasts are specialized in mineralized tissue degradation, with the combined ability to solubilize hydroxyapatite and to degrade extracellular matrix proteins. We reported previously that osteoclasts lacking Dock5 could not degrade bone due to abnormal podosome organization and absence of sealing zone formation. Consequently, adult Dock5(-/-) mice have increased trabecular bone mass. We used Dock5(-/-) mice to further investigate the different functions of osteoclast during endochondral bone formation. We show that long bones are overall morphologically normal in developing and growing Dock5(-/-) mice. We demonstrate that Dock5(-/-) mice also have normal hypertrophic cartilage and cartilage trabecular network. Conversely, trabecular bone volume increased progressively in the secondary spongiosa of Dock5(-/-) growing mice as compared to Dock5(+/+) animals, even though their osteoclast numbers were the same. In vitro, we show that Dock5(-/-) osteoclasts do present acidic compartments at the ventral plasma membrane and produce normal amounts of active MMP9, TRAP and CtsK for matrix protein degradation but they are unable to solubilize minerals. These observations reveal that contrarily to bone resorption, the ability of osteoclasts to dissolve minerals is dispensable for the degradation of mineralized hypertrophic cartilage during endochondral bone formation.
Collapse
Affiliation(s)
- Heiani Touaitahuata
- Centre de Recherche de Biochimie Macromoléculaire, CNRS UMR 5237, 1919 route de Mende, 34295 Montpellier, France; Montpellier University, France
| | - Gaelle Cres
- Centre de Recherche de Biochimie Macromoléculaire, CNRS UMR 5237, 1919 route de Mende, 34295 Montpellier, France; Montpellier University, France
| | | | - Virginie Vives
- Centre de Recherche de Biochimie Macromoléculaire, CNRS UMR 5237, 1919 route de Mende, 34295 Montpellier, France; Montpellier University, France
| | - Anne Blangy
- Centre de Recherche de Biochimie Macromoléculaire, CNRS UMR 5237, 1919 route de Mende, 34295 Montpellier, France; Montpellier University, France.
| |
Collapse
|
23
|
Barvencik F, Kurth I, Koehne T, Stauber T, Zustin J, Tsiakas K, Ludwig CF, Beil FT, Pestka JM, Hahn M, Santer R, Supanchart C, Kornak U, Del Fattore A, Jentsch TJ, Teti A, Schulz A, Schinke T, Amling M. CLCN7 and TCIRG1 mutations differentially affect bone matrix mineralization in osteopetrotic individuals. J Bone Miner Res 2014; 29:982-91. [PMID: 24108692 DOI: 10.1002/jbmr.2100] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 09/12/2013] [Accepted: 09/16/2013] [Indexed: 01/10/2023]
Abstract
Osteopetrosis is an inherited disorder of impaired bone resorption, with the most commonly affected genes being CLCN7 and TCIRG1, encoding the Cl(-) /H(+) exchanger CLC-7 and the a3 subunit of the vacuolar H(+) -ATPase, respectively. We and others have previously shown that the disease is frequently accompanied by osteomalacia, and that this additional pathology is also found in Tcirg1-deficient oc/oc mice. The remaining question was whether osteoid enrichment is specifically associated with TCIRG1 inactivation, or whether CLCN7 mutations would also cause skeletal mineralization defects. Here we describe a complete osteologic assessment of one family carrying a novel mutation in CLCN7 (D145G), which impairs the activation and relaxation kinetics of the CLC-7 ion transporter. The two siblings carrying the mutation in the homozygous state displayed high bone mass, increased serum levels of bone formation markers, but no impairment of calcium homeostasis when compared to the other family members. Most importantly, however, undecalcified processing of an iliac crest biopsy from one of the affected children clearly demonstrated a pathological increase of trabecular bone mass, but no signs of osteomalacia. Given the potential relevance of these findings we additionally performed undecalcified histology of iliac crest biopsies from seven additional cases with osteopetrosis caused by a mutation in TNFRSF11A (n=1), CLCN7 (n=3), or TCIRG1 (n=3). Here we observed that all cases with TCIRG1-dependent osteopetrosis displayed severe osteoid accumulation and decreased calcium content within the mineralized matrix. In contrast, there was no detectable bone mineralization defect in the cases with TNFRSF11A-dependent or CLCN7-dependent osteopetrosis. Taken together, our analysis demonstrates that CLCN7 and TCIRG1 mutations differentially affect bone matrix mineralization, and that there is a need to modify the current classification of osteopetrosis.
Collapse
Affiliation(s)
- Florian Barvencik
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Supanchart C, Wartosch L, Schlack C, Kühnisch J, Felsenberg D, Fuhrmann JC, de Vernejoul MC, Jentsch TJ, Kornak U. ClC-7 expression levels critically regulate bone turnover, but not gastric acid secretion. Bone 2014; 58:92-102. [PMID: 24103576 DOI: 10.1016/j.bone.2013.09.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 08/16/2013] [Accepted: 09/27/2013] [Indexed: 12/17/2022]
Abstract
Mutations in the 2Cl(-)/1H(+)-exchanger ClC-7 impair osteoclast function and cause different types of osteoclast-rich osteopetrosis. However, it is unknown to what extent ClC-7 function has to be reduced to become rate-limiting for bone resorption. In osteoclasts from osteopetrosis patients expression of the mutated ClC-7 protein did not correlate with disease severity and resorption impairment. Therefore, a series of transgenic mice expressing ClC-7 in osteoclasts at different levels was generated. Crossing of these mice with Clcn7(-/-) mutants rescued the osteopetrotic phenotype to variable degrees. One resulting double transgenic line mimicked human autosomal dominant osteopetrosis. The trabecular bone of these mice showed a reduction of osteoblast numbers, osteoid, and osteoblast marker gene expression indicative of reduced osteoblast function. In osteoclasts from these mutants ClC-7 expression levels were 20 to 30% of wildtype levels. These reduced levels not only impaired resorptive activity, but also increased numbers, size and nucleus numbers of osteoclasts differentiated in vitro. Although ClC-7 was expressed in the stomach and PTH levels were high in Clcn7(-/-) mutants loss of ClC-7 did not entail a relevant elevation of gastric pH. In conclusion, we show that in our model a reduction of ClC-7 function by approximately 70% is sufficient to increase bone mass, but does not necessarily enhance bone formation. ClC-7 does not appear to be crucially involved in gastric acid secretion, which explains the absence of an osteopetrorickets phenotype in CLCN7-related osteopetrosis.
Collapse
Affiliation(s)
- C Supanchart
- Institute of Medical Genetics and Human Genetics, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Stauber T, Weinert S, Jentsch TJ. Cell biology and physiology of CLC chloride channels and transporters. Compr Physiol 2013; 2:1701-44. [PMID: 23723021 DOI: 10.1002/cphy.c110038] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Proteins of the CLC gene family assemble to homo- or sometimes heterodimers and either function as Cl(-) channels or as Cl(-)/H(+)-exchangers. CLC proteins are present in all phyla. Detailed structural information is available from crystal structures of bacterial and algal CLCs. Mammals express nine CLC genes, four of which encode Cl(-) channels and five 2Cl(-)/H(+)-exchangers. Two accessory β-subunits are known: (1) barttin and (2) Ostm1. ClC-Ka and ClC-Kb Cl(-) channels need barttin, whereas Ostm1 is required for the function of the lysosomal ClC-7 2Cl(-)/H(+)-exchanger. ClC-1, -2, -Ka and -Kb Cl(-) channels reside in the plasma membrane and function in the control of electrical excitability of muscles or neurons, in extra- and intracellular ion homeostasis, and in transepithelial transport. The mainly endosomal/lysosomal Cl(-)/H(+)-exchangers ClC-3 to ClC-7 may facilitate vesicular acidification by shunting currents of proton pumps and increase vesicular Cl(-) concentration. ClC-3 is also present on synaptic vesicles, whereas ClC-4 and -5 can reach the plasma membrane to some extent. ClC-7/Ostm1 is coinserted with the vesicular H(+)-ATPase into the acid-secreting ruffled border membrane of osteoclasts. Mice or humans lacking ClC-7 or Ostm1 display osteopetrosis and lysosomal storage disease. Disruption of the endosomal ClC-5 Cl(-)/H(+)-exchanger leads to proteinuria and Dent's disease. Mouse models in which ClC-5 or ClC-7 is converted to uncoupled Cl(-) conductors suggest an important role of vesicular Cl(-) accumulation in these pathologies. The important functions of CLC Cl(-) channels were also revealed by human diseases and mouse models, with phenotypes including myotonia, renal loss of salt and water, deafness, blindness, leukodystrophy, and male infertility.
Collapse
Affiliation(s)
- Tobias Stauber
- Leibniz-Institut für Molekulare Pharmakologie FMP and Max-Delbrück-Centrum für Molekulare Medizin MDC, Berlin, Germany
| | | | | |
Collapse
|
26
|
Moscatelli I, Thudium CS, Flores C, Schulz A, Askmyr M, Gudmann NS, Andersen NM, Porras O, Karsdal MA, Villa A, Fasth A, Henriksen K, Richter J. Lentiviral gene transfer of TCIRG1 into peripheral blood CD34(+) cells restores osteoclast function in infantile malignant osteopetrosis. Bone 2013; 57:1-9. [PMID: 23907031 DOI: 10.1016/j.bone.2013.07.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/12/2013] [Accepted: 07/17/2013] [Indexed: 12/22/2022]
Abstract
Infantile malignant osteopetrosis (IMO) is a rare, lethal, autosomal recessive disorder characterized by non-functional osteoclasts. More than 50% of the patients have mutations in the TCIRG1 gene, encoding for a subunit of the osteoclast proton pump. The aim of this study was to restore the resorptive function of IMO osteoclasts by lentiviral mediated gene transfer of the TCIRG1 cDNA. CD34(+) cells from peripheral blood of five IMO patients and from normal cord blood were transduced with lentiviral vectors expressing TCIRG1 and GFP under a SFFV promoter, expanded in culture and differentiated on bone slices to mature osteoclasts. qPCR analysis and western blot revealed increased mRNA and protein levels of TCIRG1, comparable to controls. Vector corrected IMO osteoclasts generated increased release of Ca(2+) and bone degradation product CTX-I into the media as well as increased formation of resorption pits in the bone slices, while non-corrected IMO osteoclasts failed to resorb bone. Resorption was approximately 70-80% of that of osteoclasts generated from cord blood. Furthermore, transduced CD34(+) cells successfully engrafted in NSG-mice. In conclusion we provide the first evidence of lentiviral-mediated correction of a human genetic disease affecting the osteoclastic lineage.
Collapse
Affiliation(s)
- Ilana Moscatelli
- Department of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ochotny N, Voronov I, Owen C, Aubin JE, Manolson MF. The R740S mutation in the V-ATPase a3 subunit results in osteoclast apoptosis and defective early-stage autophagy. J Cell Biochem 2013; 114:2823-33. [DOI: 10.1002/jcb.24630] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 07/22/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Noelle Ochotny
- Faculty of Dentistry; Dental Research Institute; University of Toronto; Toronto; Ontario; Canada
| | - Irina Voronov
- Faculty of Dentistry; Dental Research Institute; University of Toronto; Toronto; Ontario; Canada
| | - Celeste Owen
- Centre for Modeling Human Disease; Samuel Lunenfeld Research Institute; Mt. Sinai Hospital; Toronto; Ontario; Canada
| | | | - Morris F. Manolson
- Faculty of Dentistry; Dental Research Institute; University of Toronto; Toronto; Ontario; Canada
| |
Collapse
|
28
|
Bollerslev J, Henriksen K, Nielsen MF, Brixen K, Van Hul W. Autosomal dominant osteopetrosis revisited: lessons from recent studies. Eur J Endocrinol 2013; 169:R39-57. [PMID: 23744590 DOI: 10.1530/eje-13-0136] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Systematic studies of autosomal dominant osteopetrosis (ADO) were followed by the identification of underlying mutations giving unique possibilities to perform translational studies. What was previously designated ADO1 turned out to be a high bone mass phenotype caused by a missense mutation in the first propeller of LRP5, a region of importance for binding inhibitory proteins. Thereby, ADO1 cannot be regarded as a classical form of osteopetrosis but must now be considered a disease of LRP5 activation. ADO (Albers-Schönberg disease, or previously ADO2) is characterized by increased number of osteoclasts and a defect in the chloride transport system (ClC-7) of importance for acidification of the resorption lacuna (a form of Chloride Channel 7 Deficiency Osteopetrosis). Ex vivo studies of osteoclasts from ADO have shown that cells do form normally but have reduced resorption capacity and an expanded life span. Bone formation seems normal despite decreased osteoclast function. Uncoupling of formation from resorption makes ADO of interest for new strategies for treatment of osteoporosis. Recent studies have integrated bone metabolism in whole-body energy homeostasis. Patients with ADO may have decreased insulin levels indicating importance beyond bone metabolism. There seems to be a paradigm shift in the treatment of osteoporosis. Targeting ClC-7 might introduce a new principle of dual action. Drugs affecting ClC-7 could be antiresorptive, still allowing ongoing bone formation. Inversely, drugs affecting the inhibitory site of LRP5 might stimulate bone formation and inhibit resorption. Thereby, these studies have highlighted several intriguing treatment possibilities, employing novel modes of action, which could provide benefits to the treatment of osteoporosis.
Collapse
Affiliation(s)
- Jens Bollerslev
- Section of Specialized Endocrinology, Medical Clinic B, Rikshospitalet, Oslo University Hospital, N-0027 Oslo, Norway.
| | | | | | | | | |
Collapse
|
29
|
Monk JA, Sims NA, Dziegielewska KM, Weiss RE, Ramsay RG, Richardson SJ. Delayed development of specific thyroid hormone-regulated events in transthyretin null mice. Am J Physiol Endocrinol Metab 2013; 304:E23-31. [PMID: 23092911 PMCID: PMC3774171 DOI: 10.1152/ajpendo.00216.2012] [Citation(s) in RCA: 24] [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] [Indexed: 12/15/2022]
Abstract
Thyroid hormones (THs) are vital for normal postnatal development. Extracellular TH distributor proteins create an intravascular reservoir of THs. Transthyretin (TTR) is a TH distributor protein in the circulatory system and is the only TH distributor protein synthesized in the central nervous system. We investigated the phenotype of TTR null mice during development. Total and free 3',5',3,5-tetraiodo-L-thyronine (T(4)) and free 3',3,5-triiodo-L-thyronine (T(3)) in plasma were significantly reduced in 14-day-old (P14) TTR null mice. TTR null mice also displayed a delayed suckling-to-weaning transition, decreased muscle mass, delayed growth, and retarded longitudinal bone growth. In addition, ileums from postnatal day 0 (P0) TTR null mice displayed disordered architecture and contained fewer goblet cells than wild type. Protein concentrations in cerebrospinal fluid from P0 and P14 TTR null mice were higher than in age-matched wild-type mice. In contrast to the current literature based on analyses of adult TTR null mice, our results demonstrate that TTR has an important and nonredundant role in influencing the development of several organs.
Collapse
Affiliation(s)
- Julie A Monk
- Department of Biochemistry and Molecular Biology, Bio21 Institute, The University of Melbourne, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
30
|
Kantaputra PN, Thawanaphong S, Issarangporn W, Klangsinsirikul P, Ohazama A, Sharpe P, Supanchart C. Long-term survival in infantile malignant autosomal recessive osteopetrosis secondary to homozygous p.Arg526Gln mutation in CLCN7. Am J Med Genet A 2012; 158A:909-16. [DOI: 10.1002/ajmg.a.35264] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 12/19/2011] [Indexed: 01/23/2023]
|
31
|
Henriksen K, Flores C, Thomsen JS, Brüel AM, Thudium CS, Neutzsky-Wulff AV, Langenbach GEJ, Sims N, Askmyr M, Martin TJ, Everts V, Karsdal MA, Richter J. Dissociation of bone resorption and bone formation in adult mice with a non-functional V-ATPase in osteoclasts leads to increased bone strength. PLoS One 2011; 6:e27482. [PMID: 22087326 PMCID: PMC3210177 DOI: 10.1371/journal.pone.0027482] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 10/17/2011] [Indexed: 01/23/2023] Open
Abstract
Osteopetrosis caused by defective acid secretion by the osteoclast, is characterized by defective bone resorption, increased osteoclast numbers, while bone formation is normal or increased. In contrast the bones are of poor quality, despite this uncoupling of formation from resorption.To shed light on the effect of uncoupling in adult mice with respect to bone strength, we transplanted irradiated three-month old normal mice with hematopoietic stem cells from control or oc/oc mice, which have defective acid secretion, and followed them for 12 to 28 weeks.Engraftment levels were assessed by flow cytometry of peripheral blood. Serum samples were collected every six weeks for measurement of bone turnover markers. At termination bones were collected for µCT and mechanical testing. An engraftment level of 98% was obtained. From week 6 until termination bone resorption was significantly reduced, while the osteoclast number was increased when comparing oc/oc to controls. Bone formation was elevated at week 6, normalized at week 12, and reduced onwards. µCT and mechanical analyses of femurs and vertebrae showed increased bone volume and bone strength of cortical and trabecular bone.In conclusion, these data show that attenuation of acid secretion in adult mice leads to uncoupling and improves bone strength.
Collapse
|