1
|
Hadjiargyrou M, Kotsiopriftis M, Lauzier D, Hamdy RC, Kloen P. Activation of Wnt signaling in human fracture callus and nonunion tissues. Bone Rep 2024; 22:101780. [PMID: 39005846 PMCID: PMC11245924 DOI: 10.1016/j.bonr.2024.101780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
The Wnt signaling pathway is a key molecular process during fracture repair. Although much of what we now know about the role of this pathway in bone is derived from in vitro and animal studies, the same cannot be said about humans. As such, we hypothesized that Wnt signaling will also be a key process in humans during physiological fracture healing as well as in the development of a nonunion (hypertrophic and oligotrophic). We further hypothesized that the expression of Wnt-signaling pathway genes/proteins would exhibit a differential expression pattern between physiological fracture callus and the pathological nonunion tissues. We tested these two hypotheses by examining the mRNA levels of key Wnt-signaling related genes: ligands (WNT4, WNT10a), receptors (FZD4, LRP5, LRP6), inhibitors (DKK1, SOST) and modulators (CTNNB1 and PORCN). RNA sequencing from calluses as well as from the two nonunion tissue types, revealed that all of these genes were expressed at about the same level in these three tissue types. Further, spatial expression experiments identified the cells responsible of producing these proteins. Robust expression was detected in osteoblasts for the majority of these genes except SOST which displayed low expression, but in contrast, was mostly detected in osteocytes. Many of these genes were also expressed by callus chondrocytes as well. Taken together, these results confirm that Wnt signaling is indeed active during both human physiological fracture healing as well as in pathological nonunions.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Maria Kotsiopriftis
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, Montreal, QC H4A 0A9, Canada
| | - Dominique Lauzier
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, Montreal, QC H4A 0A9, Canada
| | - Reggie C Hamdy
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, Montreal, QC H4A 0A9, Canada
| | - Peter Kloen
- Department of Orthopedic Surgery and Sports Medicine, Amsterdam UMC, location Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Movement Sciences, (Tissue Function and Regeneration), Amsterdam, the Netherlands
| |
Collapse
|
2
|
Celik B, Leal AF, Tomatsu S. Potential Targeting Mechanisms for Bone-Directed Therapies. Int J Mol Sci 2024; 25:8339. [PMID: 39125906 PMCID: PMC11312506 DOI: 10.3390/ijms25158339] [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: 05/21/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Bone development is characterized by complex regulation mechanisms, including signal transduction and transcription factor-related pathways, glycobiological processes, cellular interactions, transportation mechanisms, and, importantly, chemical formation resulting from hydroxyapatite. Any abnormal regulation in the bone development processes causes skeletal system-related problems. To some extent, the avascularity of cartilage and bone makes drug delivery more challenging than that of soft tissues. Recent studies have implemented many novel bone-targeting approaches to overcome drawbacks. However, none of these strategies fully corrects skeletal dysfunction, particularly in growth plate-related ones. Although direct recombinant enzymes (e.g., Vimizim for Morquio, Cerezyme for Gaucher, Elaprase for Hunter, Mepsevii for Sly diseases) or hormone infusions (estrogen for osteoporosis and osteoarthritis), traditional gene delivery (e.g., direct infusion of viral or non-viral vectors with no modifications on capsid, envelope, or nanoparticles), and cell therapy strategies (healthy bone marrow or hematopoietic stem cell transplantation) partially improve bone lesions, novel delivery methods must be addressed regarding target specificity, less immunogenicity, and duration in circulation. In addition to improvements in bone delivery, potential regulation of bone development mechanisms involving receptor-regulated pathways has also been utilized. Targeted drug delivery using organic and inorganic compounds is a promising approach in mostly preclinical settings and future clinical translation. This review comprehensively summarizes the current bone-targeting strategies based on bone structure and remodeling concepts while emphasizing potential approaches for future bone-targeting systems.
Collapse
Affiliation(s)
- Betul Celik
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
| | - Andrés Felipe Leal
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Shunji Tomatsu
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1193, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19144, USA
| |
Collapse
|
3
|
An F, Song J, Chang W, Zhang J, Gao P, Wang Y, Xiao Z, Yan C. Research Progress on the Mechanism of the SFRP-Mediated Wnt Signalling Pathway Involved in Bone Metabolism in Osteoporosis. Mol Biotechnol 2024; 66:975-990. [PMID: 38194214 DOI: 10.1007/s12033-023-01018-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024]
Abstract
Osteoporosis (OP) is a metabolic bone disease linked to an elevated fracture risk, primarily stemming from disruptions in bone metabolism. Present clinical treatments for OP merely alleviate symptoms. Hence, there exists a pressing need to identify novel targets for the clinical treatment of OP. Research indicates that the Wnt signalling pathway is modulated by serum-secreted frizzled-related protein 5 (SFRP5), potentially serving as a pivotal regulator in bone metabolism disorders. Moreover, studies confirm elevated SFRP5 expression in OP, with SFRP5 overexpression leading to the downregulation of Wnt and β-catenin proteins in the Wnt signalling pathway, as well as the expression of osteogenesis-related marker molecules such as RUNX2, ALP, and OPN. Conversely, the opposite has been reported when SFRP5 is knocked out, suggesting that SFRP5 may be a key factor involved in the regulation of bone metabolism via the Wnt signalling axis. However, the molecular mechanisms underlying the action of SFRP5-induced OP have yet to be comprehensively elucidated. This review focusses on the molecular structure and function of SFRP5 and the potential molecular mechanisms of the SFRP5-mediated Wnt signalling pathway involved in bone metabolism in OP, providing reasonable evidence for the targeted therapy of SFRP5 for the prevention and treatment of OP.
Collapse
Affiliation(s)
- Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Jiayi Song
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Weirong Chang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Jie Zhang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Peng Gao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Yujie Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Zhipan Xiao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Chunlu Yan
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
4
|
Diamond KM, Burtner AE, Siddiqui D, Alvarado K, Leake S, Rolfe S, Zhang C, Kwon RY, Maga AM. Examining craniofacial variation among crispant and mutant zebrafish models of human skeletal diseases. J Anat 2023; 243:66-77. [PMID: 36858797 PMCID: PMC10273351 DOI: 10.1111/joa.13847] [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: 08/22/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 03/03/2023] Open
Abstract
Genetic diseases affecting the skeletal system present with a wide range of symptoms that make diagnosis and treatment difficult. Genome-wide association and sequencing studies have identified genes linked to human skeletal diseases. Gene editing of zebrafish models allows researchers to further examine the link between genotype and phenotype, with the long-term goal of improving diagnosis and treatment. While current automated tools enable rapid and in-depth phenotyping of the axial skeleton, characterizing the effects of mutations on the craniofacial skeleton has been more challenging. The objective of this study was to evaluate a semi-automated screening tool can be used to quantify craniofacial variations in zebrafish models using four genes that have been associated with human skeletal diseases (meox1, plod2, sost, and wnt16) as test cases. We used traditional landmarks to ground truth our dataset and pseudolandmarks to quantify variation across the 3D cranial skeleton between the groups (somatic crispant, germline mutant, and control fish). The proposed pipeline identified variation between the crispant or mutant fish and control fish for four genes. Variation in phenotypes parallel human craniofacial symptoms for two of the four genes tested. This study demonstrates the potential as well as the limitations of our pipeline as a screening tool to examine multi-dimensional phenotypes associated with the zebrafish craniofacial skeleton.
Collapse
Affiliation(s)
- Kelly M Diamond
- Department of Biology, Rhodes College, Tennessee, Memphis, USA
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Abigail E Burtner
- Department of Biology, University of Washington, Seattle, Washington, USA
| | - Daanya Siddiqui
- Department of Biology, University of Washington, Seattle, Washington, USA
| | - Kurtis Alvarado
- Department of Biology, University of Washington, Seattle, Washington, USA
| | - Sanford Leake
- Department of Biology, University of Washington, Seattle, Washington, USA
| | - Sara Rolfe
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Chi Zhang
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Ronald Young Kwon
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - A Murat Maga
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
- Division of Craniofacial Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
5
|
Simic MK, Mohanty ST, Xiao Y, Cheng TL, Taylor VE, Charlat O, Croucher PI, McDonald MM. Multi-Targeting DKK1 and LRP6 Prevents Bone Loss and Improves Fracture Resistance in Multiple Myeloma. J Bone Miner Res 2023; 38:814-828. [PMID: 36987921 PMCID: PMC10947379 DOI: 10.1002/jbmr.4809] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 03/01/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
An imbalance between bone resorption and bone formation underlies the devastating osteolytic lesions and subsequent fractures seen in more than 90% of multiple myeloma (MM) patients. Currently, Wnt-targeted therapeutic agents that prevent soluble antagonists of the Wnt signaling pathway, sclerostin (SOST) and dickkopf-1 (DKK1), have been shown to prevent bone loss and improve bone strength in preclinical models of MM. In this study, we show increasing Wnt signaling via a novel anti-low-density lipoprotein receptor-related protein 6 (LRP6) antibody, which potentiates Wnt1-class ligand signaling through binding the Wnt receptor LRP6, prevented the development of myeloma-induced bone loss primarily through preventing bone resorption. When combined with an agent targeting the soluble Wnt antagonist DKK1, we showed more robust improvements in bone structure than anti-LRP6 treatment alone. Micro-computed tomography (μCT) analysis demonstrated substantial increases in trabecular bone volume in naïve mice given the anti-LRP6/DKK1 combination treatment strategy compared to control agents. Mice injected with 5TGM1eGFP murine myeloma cells had significant reductions in trabecular bone volume compared to naïve controls. The anti-LRP6/DKK1 combination strategy significantly improved bone volume in 5TGM1-bearing mice by 111%, which was also superior to anti-LRP6 single treatment; with similar bone structural changes observed within L4 lumbar vertebrae. Consequently, this combination strategy significantly improved resistance to fracture in lumbar vertebrae in 5TGM1-bearing mice compared to their controls, providing greater protection against fracture compared to anti-LRP6 antibody alone. Interestingly, these improvements in bone volume were primarily due to reduced bone resorption, with significant reductions in osteoclast numbers and osteoclast surface per bone surface demonstrated in 5TGM1-bearing mice treated with the anti-LRP6/DKK1 combination strategy. Importantly, Wnt stimulation with either single or combined Wnt-targeted agents did not exacerbate tumor activity. This work provides a novel approach of targeting both membrane-bound and soluble Wnt pathway components to provide superior skeletal outcomes in patients with multiple myeloma and other bone destructive cancers. © 2023 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)
- Marija K. Simic
- Skeletal Diseases ProgramGarvan Institute of Medical ResearchDarlinghurstNSWAustralia
- St Vincent's Clinical Campus, School of Clinical MedicineUniversity of New South WalesKensingtonNSWAustralia
| | - Sindhu T. Mohanty
- Skeletal Diseases ProgramGarvan Institute of Medical ResearchDarlinghurstNSWAustralia
| | - Ya Xiao
- Skeletal Diseases ProgramGarvan Institute of Medical ResearchDarlinghurstNSWAustralia
| | - Tegan L. Cheng
- Centre for Children's Bone and Musculoskeletal HealthThe Children's Hospital at WestmeadWestmeadNSWAustralia
| | - Victoria E. Taylor
- Skeletal Diseases ProgramGarvan Institute of Medical ResearchDarlinghurstNSWAustralia
| | - Olga Charlat
- Novartis Institutes for Biomedical ResearchCambridgeMAUSA
| | - Peter I. Croucher
- Skeletal Diseases ProgramGarvan Institute of Medical ResearchDarlinghurstNSWAustralia
- St Vincent's Clinical Campus, School of Clinical MedicineUniversity of New South WalesKensingtonNSWAustralia
| | - Michelle M. McDonald
- Skeletal Diseases ProgramGarvan Institute of Medical ResearchDarlinghurstNSWAustralia
- St Vincent's Clinical Campus, School of Clinical MedicineUniversity of New South WalesKensingtonNSWAustralia
- School of Medical Science, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
| |
Collapse
|
6
|
Thakur AK, Miller SE, Liau NPD, Hwang S, Hansen S, de Sousa E Melo F, Sudhamsu J, Hannoush RN. Synthetic Multivalent Disulfide-Constrained Peptide Agonists Potentiate Wnt1/β-Catenin Signaling via LRP6 Coreceptor Clustering. ACS Chem Biol 2023; 18:772-784. [PMID: 36893429 DOI: 10.1021/acschembio.2c00753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Wnt ligands are critical for tissue homeostasis and form a complex with LRP6 and frizzled coreceptors to initiate Wnt/β-catenin signaling. Yet, how different Wnts achieve various levels of signaling activation through distinct domains on LRP6 remains elusive. Developing tool ligands that target individual LRP6 domains could help elucidate the mechanism of Wnt signaling regulation and uncover pharmacological approaches for pathway modulation. We employed directed evolution of a disulfide constrained peptide (DCP) to identify molecules that bind to the third β-propeller domain of LRP6. The DCPs antagonize Wnt3a while sparing Wnt1 signaling. Using PEG linkers with different geometries, we converted the Wnt3a antagonist DCPs to multivalent molecules that potentiated Wnt1 signaling by clustering the LRP6 coreceptor. The mechanism of potentiation is unique as it occurred only in the presence of extracellular secreted Wnt1 ligand. While all DCPs recognized a similar binding interface on LRP6, they displayed different spatial orientations that influenced their cellular activities. Moreover, structural analyses revealed that the DCPs exhibited new folds that were distinct from the parent DCP framework they were evolved from. The multivalent ligand design principles highlighted in this study provide a path for developing peptide agonists that modulate different branches of cellular Wnt signaling.
Collapse
Affiliation(s)
- Avinash K Thakur
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Stephen E Miller
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Nicholas P D Liau
- Department of Structural Biology, Genentech, South San Francisco, California 94080, United States
| | - Sunhee Hwang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Simon Hansen
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| | - Felipe de Sousa E Melo
- Department of Molecular Oncology, Genentech, South San Francisco, California 94080, United States
| | - Jawahar Sudhamsu
- Department of Structural Biology, Genentech, South San Francisco, California 94080, United States
| | - Rami N Hannoush
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California 94080, United States
| |
Collapse
|
7
|
Xu C, Ji G, Chen X, Yan L, Liang T, Liu J, Wang F. Sclerostin antibody promotes bone formation through the Wnt/β-catenin signaling pathway in femoral trochlear after patellar instability. Connect Tissue Res 2023; 64:148-160. [PMID: 36379907 DOI: 10.1080/03008207.2022.2135507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE The molecular mechanism of patellar instability (PI) remains unknown. The purpose of this study was to explore the function of SOST/sclerostin in PI and examine the effect of sclerostin antibody (Scl-Ab). MATERIALS AND METHODS We randomly divided 60 male 3-week-old C57Bl/6 mice into four groups: sham, PI, Scl-Ab intraperitoneal injection (Scl-Ab IP), Scl-Ab intraarticular injection (Scl-Ab IA). PI was established in the latter three groups. The Scl-Ab IP/IA groups were administered with an intraperitoneal/intraarticular Scl-Ab injection (100 mg/kg, 20 µl), respectively, at 5-day intervals. Distal femurs were collected 30 days after the surgery. The SOST/sclerostin, β-catenin, ALP, OPG and RANKL expression in distal femur were determined. Trochlear morphology and structural parameters of the trabecular and cortical bone compartments were determined by micro-CT. Further sub-regional analysis was performed. HE staining and Masson's trichrome staining were performed to evaluate cartilage changes. RESULTS PI increased the expression of SOST/sclerostin and RANKL, and decreased β-catenin, ALP and OPG levels, while Scl-Ab IP reversed these changes. Scl-Ab IP brought trochlear morphology closer to normality. Additionally, Scl-Ab IP significantly improved most of the bone parameters. Importantly, both PI and Scl-Ab IP acted mainly on trabecular bone. Histological analysis showed that Scl-Ab IP protected cartilage from degeneration. However, Scl-Ab IA did not protect against bone loss or cartilage degradation. CONCLUSIONS SOST/sclerostin plays an important role in PI and systemic Scl-Ab use promotes bone formation through the Wnt/β-catenin signaling pathway in the femoral trochlear after PI.
Collapse
Affiliation(s)
- Chenyue Xu
- Department of Orthopaedic Surgery, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, China
| | - Gang Ji
- Department of Orthopaedic Surgery, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, China
| | - Xiaobo Chen
- Department of Orthopaedic Surgery, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, China
| | - Lirong Yan
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tuwan Liang
- College of Medical, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Junle Liu
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fei Wang
- Department of Orthopaedic Surgery, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
8
|
Craig SEL, Michalski MN, Williams BO. Got WNTS? Insight into bone health from a WNT perspective. Curr Top Dev Biol 2023; 153:327-346. [PMID: 36967199 DOI: 10.1016/bs.ctdb.2023.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
WNT signaling, essential for many aspects of development, is among the most commonly altered pathways associated with human disease. While initially studied in cancer, dysregulation of WNT signaling has been determined to be essential for skeletal development and the maintenance of bone health throughout life. In this review, we discuss the role of Wnt signaling in bone development and disease with a particular focus on two areas. First, we discuss the roles of WNT signaling pathways in skeletal development, with an emphasis on congenital and idiopathic skeletal syndromes and diseases that are associated with genetic variations in WNT signaling components. Next, we cover a topic that has long been an interest of our laboratory, how high and low levels of WNT signaling affects the establishment and maintenance of healthy bone mass. We conclude with a discussion of the status of WNT-based therapeutics in the treatment of skeletal disease.
Collapse
Affiliation(s)
- Sonya E L Craig
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, United States
| | - Megan N Michalski
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, United States
| | - Bart O Williams
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, United States.
| |
Collapse
|
9
|
Zhao D, Hua R, Riquelme MA, Cheng H, Guda T, Xu H, Gu S, Jiang JX. Osteocytes regulate bone anabolic response to mechanical loading in male mice via activation of integrin α5. Bone Res 2022; 10:49. [PMID: 35851577 PMCID: PMC9293884 DOI: 10.1038/s41413-022-00222-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/25/2022] [Accepted: 06/13/2022] [Indexed: 12/18/2022] Open
Abstract
Physical mechanical stimulation can maintain and even increase bone mass. Here, we report an important role of osteocytic integrin α5 in regulating the anabolic response of bone to mechanical loading using an Itga5 conditional gene knockout (cKO) mouse model. Integrin α5 gene deletion increased apoptotic osteocytes and reduced cortical anabolic responses to tibial compression including decreased endosteal osteoblasts and bone formation, and increased endosteal osteoclasts and bone resorption, contributing to the decreased bone area fraction and biomechanical properties, leading to an enlarged bone marrow area in cKO mice. Similar disruption of anabolic responses to mechanical loading was also detected in cKO trabecular bone. Moreover, integrin α5 deficiency impeded load-induced Cx43 hemichannel opening, and production and release of PGE2, an anabolic factor, resulting in attenuated effects of the loading on catabolic sclerostin (SOST) reduction and anabolic β-catenin increase. Together, this study shows an indispensable role of integrin α5 in osteocytes in the anabolic action of mechanical loading on skeletal tissue through activation of hemichannels and PGE2-evoked gene expression. Integrin α5 could act as a potential new therapeutic target for bone loss, especially in the elderly population with impeded mechanical sensitivity.
Collapse
Affiliation(s)
- Dezhi Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Rui Hua
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Hongyun Cheng
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas, San Antonio, TX, USA
| | - Huiyun Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
10
|
Kaya S, Schurman CA, Dole NS, Evans DS, Alliston T. Prioritization of Genes Relevant to Bone Fragility Through the Unbiased Integration of Aging Mouse Bone Transcriptomics and Human GWAS Analyses. J Bone Miner Res 2022; 37:804-817. [PMID: 35094432 PMCID: PMC9018503 DOI: 10.1002/jbmr.4516] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 12/20/2021] [Accepted: 01/17/2022] [Indexed: 11/10/2022]
Abstract
Identifying new genetic determinants of bone mineral density (BMD) and fracture promises to yield improved diagnostics and therapies for bone fragility. However, prioritizing candidate genes from genome-wide screens can be challenging. To overcome this challenge, we prioritized mouse genes that are differentially expressed in aging mouse bone based on whether their human homolog is associated with human BMD and/or fracture. Unbiased RNA-seq analysis of young and old male C57BL/6 mouse cortical bone identified 1499, 1685, and 5525 differentially expressed genes (DEGs) in 1, 2, and 2.5-year-old bone, relative to 2-month-old bone, respectively. Gene-based scores for heel ultrasound bone mineral density (eBMD) and fracture were estimated using published genome-wide association studies (GWAS) results of these traits in the UK Biobank. Enrichment analysis showed that mouse bone DEG sets for all three age groups, relative to young bone, are significantly enriched for eBMD, but only the oldest two DEG sets are enriched for fracture. Using gene-based scores, this approach prioritizes among thousands of DEGs by a factor of 5- to 100-fold, yielding 10 and 21 genes significantly associated with fracture in the two oldest groups of mouse DEGs. Though these genes were not the most differentially expressed, they included Sost, Lrp5, and others with well-established functions in bone. Several others have, as yet, unknown roles in the skeleton. Therefore, this study accelerates identification of new genetic determinants of bone fragility by prioritizing a clinically relevant and experimentally tractable number of candidate genes for functional analysis. Finally, we provide a website (www.mouse2human.org) to enable other researchers to easily apply our strategy. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
| | - Charles A. Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA
| | - Neha S. Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA
| |
Collapse
|
11
|
X-Linked Osteogenesis Imperfecta Possibly Caused by a Novel Variant in PLS3. Genes (Basel) 2021; 12:genes12121851. [PMID: 34946798 PMCID: PMC8701009 DOI: 10.3390/genes12121851] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/11/2022] Open
Abstract
Osteogenesis imperfecta (OI) represents a complex spectrum of genetic bone diseases that occur primarily due to mutations and deletions of the COL1A1 and COL1A2 genes. Recent molecular studies of the network of signaling pathways have contributed to a better understanding of bone remodeling and the pathogenesis of OI caused by mutations in many other genes associated with normal bone mineralization. In this paper, a case of a rare X-linked variant of OI with a change in the gene encoding plastin 3—a protein important for the regulation of the actin cytoskeleton, is presented. A 16-year-old patient developed ten bone fractures caused by minor trauma or injury, including a compression fracture of the second lumbar vertebra during his lifetime. Next-generation sequencing analysis did not show pathologically relevant deviations in the COL1A1 and COL1A2 genes. Targeted gene analyses (Skeletal disorder panel) of the patient, his father, mother and sister were then performed, detecting variants of uncertain significance (VUS) for genes PLS3, FN1 and COL11A2. A variant in the PLS3 gene were identified in the patient, his mother and sister. Since the PLS3 gene is located on the X chromosome, the mother and sister showed no signs of the disease. Although the variant in the PLS3 gene (c.685G>A (p.Gly229Arg)) has not yet been described in the literature, nor is its pathogenicity known, clinical findings combined with genetic testing showed that this variant may explain the cause of X-linked OI in our patient. This rare case of the PLS3 variant of X-linked OI might point to a novel target for personalized therapy in patients with this severe disease.
Collapse
|
12
|
Shimohira T, Niimi H, Ohsugi Y, Tsuchiya Y, Morita K, Yoshida S, Hatasa M, Shiba T, Kadokura H, Yokose S, Katagiri S, Iwata T, Aoki A. Low-Level Erbium-Doped Yttrium Aluminum Garnet Laser Irradiation Induced Alteration of Gene Expression in Osteogenic Cells from Rat Calvariae. PHOTOBIOMODULATION PHOTOMEDICINE AND LASER SURGERY 2021; 39:566-577. [PMID: 34339325 DOI: 10.1089/photob.2020.4958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Objective: The aim of this study was to investigate the effect of low-level erbium-doped yttrium aluminum garnet (Er:YAG) laser irradiation on gene expression in osteogenic cells from rat calvariae. Background: Previous studies showed beneficial effects of laser irradiation on bone-related cells. However, few studies have examined the gene expression alteration by laser irradiation on osteogenic cells in a calcified condition. Materials and methods: Osteogenic cells were prepared by culturing rat calvarial osteoblast-like cells in osteoinductive medium for 21 days. The cells at the bottom of the culture dish were irradiated with Er:YAG laser (wavelength: 2.94 μm, energy density: 3.1 and 8.2 J/cm2) positioned at distance of 25 cm. Lactate dehydrogenase (LDH) assay of the irradiated cells was performed. After screening for genes related to bone formation, mechanotransduction, and thermal effect by quantitative polymerase chain reaction (qPCR), gene expression at 3 h after 3.1 J/cm2 irradiation was comprehensively analyzed using microarray. Results: No dramatical increase in surface temperature and LDH activities after laser irradiation were observed. Sost expression was significantly reduced at 3 h after 3.1 J/cm2 irradiation. Bcar1 and Hspa1a expression was significantly increased following 8.2 J/cm2 irradiation. Microarray analysis identified 116 differentially expressed genes. Gene set enrichment analysis showed enrichment of histone H3-K9 methylation and modification gene sets. Conclusions: Er:YAG laser irradiation, especially at 3.1 J/cm2, showed positive effect on the expression of genes related to bone formation in osteogenic cells, without inducing significant cell damage. These findings may represent critical mechanisms of early bone formation after Er:YAG laser irradiation.
Collapse
Affiliation(s)
- Tsuyoshi Shimohira
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiromi Niimi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yujin Ohsugi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yosuke Tsuchiya
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kazuki Morita
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Sumiko Yoshida
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masahiro Hatasa
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takahiko Shiba
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroshi Kadokura
- Division of Endodontic and Operative Dentistry, Department of Restorative and Biomaterials Sciences, School of Dentistry, Meikai University, Saitama, Japan
| | - Satoshi Yokose
- Division of Endodontic and Operative Dentistry, Department of Restorative and Biomaterials Sciences, School of Dentistry, Meikai University, Saitama, Japan
| | - Sayaka Katagiri
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Akira Aoki
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
13
|
Costa S, Fairfield H, Farrell M, Murphy CS, Soucy A, Vary C, Holdsworth G, Reagan MR. Sclerostin antibody increases trabecular bone and bone mechanical properties by increasing osteoblast activity damaged by whole-body irradiation in mice. Bone 2021; 147:115918. [PMID: 33737193 PMCID: PMC8076093 DOI: 10.1016/j.bone.2021.115918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/22/2021] [Accepted: 03/11/2021] [Indexed: 12/16/2022]
Abstract
Irradiation therapy causes bone deterioration and increased risk for skeletal-related events. Irradiation interferes with trabecular architecture through increased osteoclastic activity, decreased osteoblastic activity, and increased adipocyte expansion in the bone marrow (BM), which further compounds bone-related disease. Neutralizing antibodies to sclerostin (Scl-Ab) increase bone mass and strength by increasing bone formation and reducing bone resorption. We hypothesized that treatment with Scl-Ab would attenuate the adverse effects of irradiation by increasing bone volume and decreasing BM adipose tissue (BMAT), resulting in better quality bone. In this study, 12-week-old female C57BL/6J mice were exposed to 6 Gy whole-body irradiation or were non-irradiated, then administered Scl-Ab (25 mg/kg) or vehicle weekly for 5 weeks. Femoral μCT analysis confirmed that the overall effect of IR significantly decreased trabecular bone volume/total volume (Tb.BV/TV) (2-way ANOVA, p < 0.0001) with a -43.8% loss in Tb.BV/TV in the IR control group. Scl-Ab independently increased Tb.BV/TV by 3.07-fold in non-irradiated and 3.6-fold in irradiated mice (2-way ANOVA, p < 0.0001). Irradiation did not affect cortical parameters, although Scl-Ab increased cortical thickness and area significantly in both irradiated and non-irradiated mice (2-way ANOVA, p < 0.0001). Femoral mechanical testing confirmed Scl-Ab significantly increased bending rigidity and ultimate moment independently of irradiation (2-way ANOVA, p < 0.0001). Static and dynamic histomorphometry of the femoral metaphysis revealed osteoblast vigor, not number, was significantly increased in the irradiated mice treated with Scl-Ab. Systemic alterations were assessed through serum lipidomic analysis, which showed that Scl-Ab normalized lipid profiles in the irradiated group. This data supports the theory of sclerostin as a novel contributor to the regulation of osteoblast activity after irradiation. Overall, our data support the hypothesis that Scl-Ab ameliorates the deleterious effects of whole-body irradiation on bone and adipose tissue in a mouse model. Our findings suggest that future research into localized and systemic therapies after irradiation exposure is warranted.
Collapse
Affiliation(s)
- Samantha Costa
- Maine Medical Center Research Institute, Scarborough, ME, USA; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Heather Fairfield
- Maine Medical Center Research Institute, Scarborough, ME, USA; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Mariah Farrell
- Maine Medical Center Research Institute, Scarborough, ME, USA; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Connor S Murphy
- Maine Medical Center Research Institute, Scarborough, ME, USA; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Ashley Soucy
- Maine Medical Center Research Institute, Scarborough, ME, USA; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA
| | - Calvin Vary
- Maine Medical Center Research Institute, Scarborough, ME, USA; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA; Tufts University School of Medicine, Boston, MA, USA
| | | | - Michaela R Reagan
- Maine Medical Center Research Institute, Scarborough, ME, USA; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA; Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
14
|
Shams R, Drasites KP, Zaman V, Matzelle D, Shields DC, Garner DP, Sole CJ, Haque A, Banik NL. The Pathophysiology of Osteoporosis after Spinal Cord Injury. Int J Mol Sci 2021; 22:3057. [PMID: 33802713 PMCID: PMC8002377 DOI: 10.3390/ijms22063057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) affects approximately 300,000 people in the United States. Most individuals who sustain severe SCI also develop subsequent osteoporosis. However, beyond immobilization-related lack of long bone loading, multiple mechanisms of SCI-related bone density loss are incompletely understood. Recent findings suggest neuronal impairment and disability may lead to an upregulation of receptor activator of nuclear factor-κB ligand (RANKL), which promotes bone resorption. Disruption of Wnt signaling and dysregulation of RANKL may also contribute to the pathogenesis of SCI-related osteoporosis. Estrogenic effects may protect bones from resorption by decreasing the upregulation of RANKL. This review will discuss the current proposed physiological and cellular mechanisms explaining osteoporosis associated with SCI. In addition, we will discuss emerging pharmacological and physiological treatment strategies, including the promising effects of estrogen on cellular protection.
Collapse
Affiliation(s)
- Ramsha Shams
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Kelsey P. Drasites
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Vandana Zaman
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St., Charleston, SC 29401, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St., Charleston, SC 29401, USA
| | - Donald C. Shields
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
| | - Dena P. Garner
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Christopher J. Sole
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Narendra L. Banik
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St., Charleston, SC 29401, USA
| |
Collapse
|
15
|
Wang F, Rummukainen P, Heino TJ, Kiviranta R. Osteoblastic Wnt1 regulates periosteal bone formation in adult mice. Bone 2021; 143:115754. [PMID: 33189914 DOI: 10.1016/j.bone.2020.115754] [Citation(s) in RCA: 6] [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: 05/28/2020] [Revised: 10/25/2020] [Accepted: 11/10/2020] [Indexed: 10/23/2022]
Abstract
Compelling clinical data together with genetically modified mouse models have demonstrated that Wnt1 is a key Wnt ligand in bone metabolism, regulating both osteoblast activity and osteoclast differentiation. We have previously shown that deletion of Wnt1 in limb mesenchymal cells leads to severe ostepenic bone phenotype and spontaneous fractures very early after birth. However, the function of Wnt1 in mature skeleton remained unknown. To investigate the role of Wnt1 specifically in adult bone metabolism, we generated an osteoblast lineage-targeted inducible Wnt1 knockout mouse model using tetracycline-controlled Osterix-Cre mouse line (Osx-Cre). In this model, the Cre recombinase expression is suppressed by administering doxycycline (Dox) in drinking water. As expected, Wnt1Osx-/- mice without Dox developed spontaneous fractures early by 3 weeks of age due to severe trabecular and cortical osteopenia. Administration of Dox to Wnt1Osx-Dox-/- and control mice until 4 weeks of age suppressed Wnt1 deletion and completely prevented the fractures. Withdrawal of Dox led to deletion in Wnt1 allele but the fracture incidence progressively decreased in Wnt1Osx-Dox-/- mice at 8 or 12 weeks of age (4 and 8 weeks after Dox withdrawal). Interestingly, deletion of Wnt1 at 4 weeks of age resulted only in a modest and transient trabecular osteopenia that was more pronounced in females and was normalized by 12 weeks of age. However, diaphyseal cortical bone mass and cortical thickness in the femurs were significantly decreased in Wnt1Osx-Dox-/- mice of both genders. Mechanisticly, this was due to impaired periosteal bone formation. Based on our data, in addition to its essential role in early skeletal growth, Wnt1 is an important regulator of modeling-based bone formation and cortical thickness in adult mice.
Collapse
Affiliation(s)
- Fan Wang
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Terhi J Heino
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Riku Kiviranta
- Institute of Biomedicine, University of Turku, Turku, Finland; Department of Endocrinology, Division of Medicine, University of Turku and Turku University Hospital, Turku, Finland.
| |
Collapse
|
16
|
Comprehensive and Sequential Gene Expression Analysis of Bone Healing Process Following Er:YAG Laser Ablation. PHOTOBIOMODULATION PHOTOMEDICINE AND LASER SURGERY 2021; 39:100-112. [DOI: 10.1089/photob.2020.4833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Lim KE, Bullock WA, Horan DJ, Williams BO, Warman ML, Robling AG. Co-deletion of Lrp5 and Lrp6 in the skeleton severely diminishes bone gain from sclerostin antibody administration. Bone 2021; 143:115708. [PMID: 33164872 PMCID: PMC7770084 DOI: 10.1016/j.bone.2020.115708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 10/01/2020] [Accepted: 10/20/2020] [Indexed: 01/14/2023]
Abstract
The cysteine knot protein sclerostin is an osteocyte-derived secreted inhibitor of the Wnt co-receptors LRP5 and LRP6. LRP5 plays a dominant role in bone homeostasis, but we previously reported that Sost/sclerostin suppression significantly increased osteogenesis regardless of Lrp5 presence or absence. Those observations suggested that the bone forming effects of sclerostin inhibition can occur through Lrp6 (when Lrp5 is suppressed), or through other yet undiscovered mechanisms independent of Lrp5/6. To distinguish between these two possibilities, we generated mice with compound deletion of Lrp5 and Lrp6 selectively in bone, and treated them with sclerostin monoclonal antibody (Scl-mAb). All mice were homozygous flox for both Lrp5 and Lrp6 (Lrp5f/f; Lrp6f/f), and varied only in whether or not they carried the Dmp1-Cre transgene. Positive (Cre+) and negative (Cre-) mice were injected with Scl-mAb or vehicle from 4.5 to 14 weeks of age. Vehicle-treated Cre+ mice exhibited significantly reduced skeletal properties compared to vehicle-treated Cre- mice, as assessed by DXA, μCT, pQCT, and histology, indicating that Lrp5/6 deletions were effective and efficient. Scl-mAb treatment improved nearly every bone-related parameter among Cre- mice, but the same treatment in Cre+ mice resulted in little to no improvement in skeletal properties. For the few endpoints where Cre+ mice responded to Scl-mAb, it is likely that antibody-induced promotion of Wnt signaling occurred in cell types earlier in the mesenchymal/osteoblast differentiation pathway than the Dmp1-expressing stage. This latter conclusion was supported by changes in some histomorphometric parameters. In conclusion, unlike with the deletion of Lrp5 alone, the bone-selective late-stage co-deletion of Lrp5 and Lrp6 significantly impairs or completely nullifies the osteogenic action of Scl-mAb, and highlights a major role for both Lrp5 and Lrp6 in the mechanism of action for the bone-building effects of sclerostin antibody.
Collapse
Affiliation(s)
- Kyung-Eun Lim
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Whitney A Bullock
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Daniel J Horan
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bart O Williams
- Program for Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Matthew L Warman
- Department of Orthopaedic Surgery, Boston Children's Hospital, Boston, MA, USA
| | - Alexander G Robling
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA; Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA; Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA.
| |
Collapse
|
18
|
Wang B, Guo H, Geng T, Sun K, Zhang L, Lu Z, Jin Q. The effect of strontium ranelate on titanium particle-induced periprosthetic osteolysis regulated by WNT/β-catenin signaling in vivo and in vitro. Biosci Rep 2021; 41:BSR20203003. [PMID: 33443286 PMCID: PMC7846966 DOI: 10.1042/bsr20203003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022] Open
Abstract
Aseptic loosening following periprosthetic osteolysis is the primary complication that limits the lifetime of total joint arthroplasty (TJA). The wear particles trigger a chronic inflammation response in the periprosthetic tissue and turn over the bone balance to bone resorption. The present study aimed to investigate the possible effect and mechanism of strontium ranelate (SR), a clinically safe drug for osteoporosis, on particle-induced periprosthetic osteolysis. Thirty-six female C57BL/6j mice underwent tibial Ti-nail implantation to establish an animal model of aseptic loosening. After 12 weeks, micro-CT results showed that strontium ranelate could inhibit periprosthetic bone resorption. In vitro, Ti particles were used to stimulate RAW264.7 cell line to collect conditioned medium, and co-culture MC3T3-E1 cell line with conditioned medium to establish a cell model of aseptic loosening. The results of alkaline phosphatase (ALP) detection, immunofluorescence, and flow cytometry demonstrated that strontium ranelate could regulate the expression of OPG/RANKL, promote differentiation and mineralization, and inhibit apoptosis in osteoblasts. Moreover, we revealed that SR's exerted its therapeutic effect by down-regulating sclerostin, thereby activating the Wnt/β-catenin signal pathway. Therefore, this research suggests that strontium ranelate could be a potential drug for the prevention and treatment of particle-induced aseptic loosening post-TJA.
Collapse
Affiliation(s)
- Bolun Wang
- Department of Orthopedic Surgery, Ningxia Medical University, 1160 Shengli Street, Xingqing Area, Yinchuan, Ningxia, P.R. China 750004
| | - Haohui Guo
- Department of Orthopedics, General Hospital of Ningxia Medical University, 804 Shengli Street, Xingqing Area, Yinchuan, Ningxia, P.R. China 750004
| | - Tianxiang Geng
- Department of Orthopedic Surgery, Ningxia Medical University, 1160 Shengli Street, Xingqing Area, Yinchuan, Ningxia, P.R. China 750004
| | - Kening Sun
- Department of Orthopedics, General Hospital of Ningxia Medical University, 804 Shengli Street, Xingqing Area, Yinchuan, Ningxia, P.R. China 750004
| | - Liang Zhang
- Department of Orthopedics, General Hospital of Ningxia Medical University, 804 Shengli Street, Xingqing Area, Yinchuan, Ningxia, P.R. China 750004
| | - Zhidong Lu
- Department of Orthopedics, General Hospital of Ningxia Medical University, 804 Shengli Street, Xingqing Area, Yinchuan, Ningxia, P.R. China 750004
| | - Qunhua Jin
- Department of Orthopedics, General Hospital of Ningxia Medical University, 804 Shengli Street, Xingqing Area, Yinchuan, Ningxia, P.R. China 750004
| |
Collapse
|
19
|
Li X, Lu X, Fan D, Li L, Lu C, Tan Y, Xia Y, Zhao H, Fan M, Xiao C. Synergistic Effects of Erzhi Pill Combined With Methotrexate on Osteoblasts Mediated via the Wnt1/LRP5/ β-Catenin Signaling Pathway in Collagen-Induced Arthritis Rats. Front Pharmacol 2020; 11:228. [PMID: 32218732 PMCID: PMC7079734 DOI: 10.3389/fphar.2020.00228] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/19/2020] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by chronic synovitis, bone erosion, and bone loss. Erzhi Pill (EZP), a classic Chinese patent medicine, is often used to treat osteoporosis and shows a capacity for bone metabolism regulation. Methotrexate (MTX), an essential drug for RA treatment, has been reported to inhibit generalized bone loss in RA patients. However, the combined therapeutic effects and mechanism of EZP and MTX in RA have not been fully elucidated. The aim of this study was to investigate the synergistic effect of EZP and MTX on RA and to explore the underlying mechanism through network pharmacological prediction and experimental verification. Chemical compounds of EZP, human target proteins of EZP and MTX, and RA-related human genes were identified in the Encyclopedia of Traditional Chinese Medicine database, PubChem database, and NCBI database, respectively. The molecular network of EZP and MTX in RA was generated and analyzed with Ingenuity Pathway Analysis software according to the datasets. Then, MTX monotherapy, EZP monotherapy, and combined MTX and EZP therapy were administered to collagen-induced arthritis rats, followed by assessment of pathological score, bone damage, bone alkaline phosphatases (BALP), and tartrate-resistant acid phosphatase (TRACP), and of gene levels related to the Wnt1/LRP5/β-catenin pathway according to network pharmacological analysis. Finally, serum samples from MTX-, EZP- and MTX+EZP-treated rats were used to treat the rat osteoblast (OB)-like UMR-106 cell line to evaluate gene levels related to Wnt1/LRP5/β-catenin. Network pharmacological analysis showed that the Wnt/β-catenin signaling pathway was the top signaling pathway shared among MTX, EZP, and RA. The results from in vivo experiments indicated that EZP combined with MTX reduced arthritis severity, alleviated ankle bone damage, increased BALP and decreased TRACP serum levels, and regulated the mRNA expression of Wnt1, LRP5, β-catenin, Runx2, BALP, and BGP in the ankles. In vitro experiments showed that EZP combined with MTX could also improve the expression of genes related to the Wnt1/LRP5/β-catenin pathway. This study demonstrated that EZP in combination with MTX played a synergistic role in regulating OBs in RA, which was connected to the modulatory effect of EZP and MTX on the Wnt1/LRP5/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xiaoya Li
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China.,Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Xiangcheng Lu
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Danping Fan
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong Tan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ya Xia
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hongyan Zhao
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Miaoxuan Fan
- Beijing Institute for Drug Control, NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing, China
| | - Cheng Xiao
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China.,Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Morse A, Ko FC, McDonald MM, Lee LR, Schindeler A, van der Meulen MCH, Little DG. Increased anabolic bone response in Dkk1 KO mice following tibial compressive loading. Bone 2020; 131:115054. [PMID: 31521827 DOI: 10.1016/j.bone.2019.115054] [Citation(s) in RCA: 7] [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: 05/23/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022]
Abstract
A viable Dkk1 knockout (KO) mouse strain in which embryonic lethality is rescued by developmental Wnt3 heterozygosity (Dkk1-/-:Wnt3+/-) exhibits increased bone formation and a high bone mass phenotype. We hypothesized that in vivo mechanical loading would further augment the bone formation response in Dkk1 KO mice, comparable to results from Sost KO mice. A cyclic loading protocol was applied to Dkk1 KO mice, wild type mice (WT; Dkk1+/+:Wnt3+/+), and Wnt3 heterozygote (Wnt3+/-; Dkk1+/+:Wnt3+/-) controls. The left tibiae of 10-week-old female mice were dynamically loaded in vivo with 7N maximum compressive force 5 days/week for 2 weeks. Dkk1 KO bones were significantly stiffer, and so an additional group of Dkk1 KO received 12N maximum compressive force to achieve an equivalent +1200με strain at the mid-diaphysis. MicroCT and bone histomorphometry analyses were subsequently performed. All groups responded to tibial loading with increased mid-diaphyseal bone volume. The largest effect size was in the Dkk1 KO -12N group. Thus, Dkk1 KO animals had enhanced sensitivity to mechanical loading. Increases in cortical bone volume reflected increased periosteal bone formation. Bone volume and formation were not altered between WT and Wnt3+/- controls. These data support the concept that agonists of Wnt/β-catenin signaling can act synergistically with load-bearing exercise. Notably, Sost expression decreased with loading in Dkk1 KO and WT mice, independent of genotype. These data suggest that a compensatory downregulation of Sost in Dkk1 KO mice is not likely the primary mechanism for the augmented response to mechanical load.
Collapse
Affiliation(s)
- Alyson Morse
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Frank C Ko
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Michelle M McDonald
- Healthy Aging Theme, Bone Biology, The Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia
| | - Lucinda R Lee
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Marjolein C H van der Meulen
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States; Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States; Research Division, Hospital for Special Surgery, NY, United States
| | - David G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, Australia.
| |
Collapse
|
21
|
Luther J, Yorgan TA, Rolvien T, Ulsamer L, Koehne T, Liao N, Keller D, Vollersen N, Teufel S, Neven M, Peters S, Schweizer M, Trumpp A, Rosigkeit S, Bockamp E, Mundlos S, Kornak U, Oheim R, Amling M, Schinke T, David JP. Wnt1 is an Lrp5-independent bone-anabolic Wnt ligand. Sci Transl Med 2019; 10:10/466/eaau7137. [PMID: 30404864 DOI: 10.1126/scitranslmed.aau7137] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022]
Abstract
WNT1 mutations in humans are associated with a new form of osteogenesis imperfecta and with early-onset osteoporosis, suggesting a key role of WNT1 in bone mass regulation. However, the general mode of action and the therapeutic potential of Wnt1 in clinically relevant situations such as aging remain to be established. Here, we report the high prevalence of heterozygous WNT1 mutations in patients with early-onset osteoporosis. We show that inactivation of Wnt1 in osteoblasts causes severe osteoporosis and spontaneous bone fractures in mice. In contrast, conditional Wnt1 expression in osteoblasts promoted rapid bone mass increase in developing young, adult, and aged mice by rapidly increasing osteoblast numbers and function. Contrary to current mechanistic models, loss of Lrp5, the co-receptor thought to transmit extracellular WNT signals during bone mass regulation, did not reduce the bone-anabolic effect of Wnt1, providing direct evidence that Wnt1 function does not require the LRP5 co-receptor. The identification of Wnt1 as a regulator of bone formation and remodeling provides the basis for development of Wnt1-targeting drugs for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Julia Luther
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lorenz Ulsamer
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Till Koehne
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.,Department of Orthodontics, University Medical Center Hamburg-Eppendorf, D 20246 Hamburg, Germany
| | - Nannan Liao
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Daniela Keller
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nele Vollersen
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Teufel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mona Neven
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stephanie Peters
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michaela Schweizer
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, D 20251 Hamburg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), D 69120 Heidelberg, Germany
| | - Sebastian Rosigkeit
- Institute for Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Ernesto Bockamp
- Institute for Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Stefan Mundlos
- Institute of Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, D 13353 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, D 13353 Berlin, Germany.,Max Planck Institute for Molecular Genetics, D 14195 Berlin, Germany
| | - Uwe Kornak
- Institute of Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, D 13353 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, D 13353 Berlin, Germany.,Max Planck Institute for Molecular Genetics, D 14195 Berlin, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Jean-Pierre David
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
22
|
Autosomal-Recessive Mutations in MESD Cause Osteogenesis Imperfecta. Am J Hum Genet 2019; 105:836-843. [PMID: 31564437 DOI: 10.1016/j.ajhg.2019.08.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/16/2019] [Indexed: 11/21/2022] Open
Abstract
Osteogenesis imperfecta (OI) comprises a genetically heterogeneous group of skeletal fragility diseases. Here, we report on five independent families with a progressively deforming type of OI, in whom we identified four homozygous truncation or frameshift mutations in MESD. Affected individuals had recurrent fractures and at least one had oligodontia. MESD encodes an endoplasmic reticulum (ER) chaperone protein for the canonical Wingless-related integration site (WNT) signaling receptors LRP5 and LRP6. Because complete absence of MESD causes embryonic lethality in mice, we hypothesized that the OI-associated mutations are hypomorphic alleles since these mutations occur downstream of the chaperone activity domain but upstream of ER-retention domain. This would be consistent with the clinical phenotypes of skeletal fragility and oligodontia in persons deficient for LRP5 and LRP6, respectively. When we expressed wild-type (WT) and mutant MESD in HEK293T cells, we detected WT MESD in cell lysate but not in conditioned medium, whereas the converse was true for mutant MESD. We observed that both WT and mutant MESD retained the ability to chaperone LRP5. Thus, OI-associated MESD mutations produce hypomorphic alleles whose failure to remain within the ER significantly reduces but does not completely eliminate LRP5 and LRP6 trafficking. Since these individuals have no eye abnormalities (which occur in individuals completely lacking LRP5) and have neither limb nor brain patterning defects (both of which occur in mice completely lacking LRP6), we infer that bone mass accrual and dental patterning are more sensitive to reduced canonical WNT signaling than are other developmental processes. Biologic agents that can increase LRP5 and LRP6-mediated WNT signaling could benefit individuals with MESD-associated OI.
Collapse
|
23
|
Ohsugi Y, Aoki A, Mizutani K, Katagiri S, Komaki M, Noda M, Takagi T, Kakizaki S, Meinzer W, Izumi Y. Evaluation of bone healing following Er:YAG laser ablation in rat calvaria compared with bur drilling. JOURNAL OF BIOPHOTONICS 2019; 12:e201800245. [PMID: 30324711 DOI: 10.1002/jbio.201800245] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 06/08/2023]
Abstract
The Er:YAG laser is currently used for bone ablation. However, the effect of Er:YAG laser irradiation on bone healing remains unclear. The aim of this study was to investigate bone healing following ablation by laser irradiation as compared with bur drilling. Rat calvarial bone was ablated using Er:YAG laser or bur with water coolant. Er:YAG laser effectively ablated bone without major thermal changes. In vivo micro-computed tomography analysis revealed that laser irradiation showed significantly higher bone repair ratios than bur drilling. Scanning electron microscope analysis showed more fibrin deposition on laser-ablated bone surfaces. Microarray analysis followed by gene set enrichment analysis revealed that IL6/JAK/STAT3 signaling and inflammatory response gene sets were enriched in bur-drilled bone at 6 hours, whereas the E2F targets gene set was enriched in laser-irradiated bone. Additionally, Hspa1a and Dmp1 expressions were increased and Sost expression was decreased in laser-irradiated bone compared with bur-drilled bone. In granulation tissue formed after laser ablation, Alpl and Gblap expressions increased compared to bur-drilled site. Immunohistochemistry showed that osteocalcin-positive area was increased in the laser-ablated site. These results suggest that Er:YAG laser might accelerate early new bone formation with advantageous surface changes and cellular responses for wound healing, compared with bur-drilling.
Collapse
Affiliation(s)
- Yujin Ohsugi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akira Aoki
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koji Mizutani
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sayaka Katagiri
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Motohiro Komaki
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Highly Advanced Stomatology, Graduate School of Dentistry, Kanagawa Dental University, Yokosuka, Japan
| | - Masahiro Noda
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toru Takagi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sho Kakizaki
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Walter Meinzer
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuichi Izumi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
24
|
Cao Y, Wang B, Wang D, Zhan D, Mai C, Wang P, Wei Q, Liu Y, Wang H, He W, Xu L. Expression of Sclerostin in Osteoporotic Fracture Patients Is Associated with DNA Methylation in the CpG Island of the SOST Gene. Int J Genomics 2019; 2019:7076513. [PMID: 30729116 PMCID: PMC6341240 DOI: 10.1155/2019/7076513] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/26/2018] [Accepted: 10/11/2018] [Indexed: 01/17/2023] Open
Abstract
PURPOSE SOST gene is one of the key factors in regulating bone absorption. Although there are reports showing diverse transcription factors, epigenetic modification could be responsible for regulating SOST gene expression. There is still little exploration on promoter methylation status of SOST gene in osteoporotic bone tissues. The aim of this study is to investigate the involvement of CpG methylation in regulation of SOST expression in patients with primary osteoporosis. METHODS The diagnosis of osteoporosis was established on the basis of dual energy X-ray absorptiometry to measure BMD. All femoral bone tissues were separated in surgeries. After extracting total RNA and protein, we checked the relative expression levels of SOST by quantitative real-time PCR and western blot. Also, immunohistochemical staining was performed to observe the expression of SOST protein in the bone samples. The genomic DNA of non-OPF (non-osteoporotic fracture bone tissues) and OPF (osteoporotic fracture bone tissues) were treated by bisulfite modification, and methylation status of CpG sites in the CpG island of SOST gene promoter was determined by DNA sequencing. RESULTS SOST gene expression in the non-OPF group was lower than that in OPF group. Bisulfite sequencing result showed that SOST gene promoter was slightly demethylated in the OPF group, as compared with non-OPF group. CONCLUSION Our study demonstrated that DNA methylation influenced the transcriptional expression of SOST gene, which probably may play an important role in the pathogenesis of primary osteoporosis.
Collapse
Affiliation(s)
- Yanming Cao
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bin Wang
- Department of Orthopedics, People's Hospital of Sanshui, Foshan, China
| | - Ding Wang
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dongxiang Zhan
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Caiyuan Mai
- Department of Obstetrics, Guangdong Women and Children's Hospital, Guangzhou 510010, China
| | - Peng Wang
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiushi Wei
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yamei Liu
- Departments of Diagnostics of Traditional Chinese Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Haibin Wang
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei He
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liangliang Xu
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
25
|
Morse A, Cheng TL, Schindeler A, McDonald MM, Mohanty ST, Kneissel M, Kramer I, Little DG. Dkk1 KO Mice Treated with Sclerostin Antibody Have Additional Increases in Bone Volume. Calcif Tissue Int 2018; 103:298-310. [PMID: 29845410 DOI: 10.1007/s00223-018-0420-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/05/2018] [Indexed: 12/17/2022]
Abstract
Dickkopf-1 (DKK1) and sclerostin are antagonists of the Wnt/β-catenin pathway and decreased expression of either results in increased bone formation and mass. As both affect the same signaling pathway, we aimed to elucidate the redundancy and/or compensation of sclerostin and DKK1. Weekly sclerostin antibody (Scl-Ab) was used to treat 9-week-old female Dkk1 KO (Dkk1-/-:Wnt3+/-) mice and compared to Scl-Ab-treated wild-type mice as well as vehicle-treated Dkk1 KO and wild-type animals. While Wnt3 heterozygote (Wnt3+/-) mice show no bone phenotype, Scl-Ab and vehicle-treated control groups of this genotype were included. Specimens were harvested after 3 weeks for microCT, bone histomorphometry, anti-sclerostin immunohistochemistry, and biomechanical testing. Scl-Ab enhanced bone anabolism in all treatment groups, but with synergistic enhancement seen in the cancellous compartment of Dkk1 KO mice (bone volume + 55% Dkk1 KO p < 0.01; + 22% wild type p < 0.05). Scl-Ab treatment produced less marked increases in cortical bone of the tibiae, with anabolic effects similar across genotypes. Mechanical testing confirmed that Scl-Ab improved strength across all genotypes; however, no enhancement was seen within Dkk1 KO mice. Dynamic bone labeling showed that Scl-Ab treatment was associated with increased bone formation, regardless of genotype. Immunohistochemical staining for sclerostin protein indicated no differences in the Dkk1 KO mice, indicating that the increased Wnt signaling associated with DKK1 deficiency was not compensated by upregulation of sclerostin protein. These data suggest complex interactions between Wnt signaling factors in bone, but critically illustrate synergy between DKK1 deficiency and Scl-Ab treatment. These data support the application of dual-targeted therapeutics in the modulation of bone anabolism.
Collapse
Affiliation(s)
- Alyson Morse
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
- Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Tegan L Cheng
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
- Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia
- Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Michelle M McDonald
- Bone Biology Program, The Garvan Institute of Medical Research, Sydney, Australia
| | - Sindhu T Mohanty
- Bone Biology Program, The Garvan Institute of Medical Research, Sydney, Australia
| | | | | | - David G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW, 2145, Australia.
- Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia.
| |
Collapse
|
26
|
Geng T, Sun S, Yu H, Guo H, Zheng M, Zhang S, Chen X, Jin Q. Strontium ranelate inhibits wear particle-induced aseptic loosening in mice. ACTA ACUST UNITED AC 2018; 51:e7414. [PMID: 29995108 PMCID: PMC6050946 DOI: 10.1590/1414-431x20187414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 05/17/2018] [Indexed: 11/21/2022]
Abstract
The imbalance between bone formation and osteolysis plays a key role in the pathogenesis of aseptic loosening. Strontium ranelate (SR) can promote bone formation and inhibit osteolysis. The aim of this study was to explore the role and mechanism of SR in aseptic loosening induced by wear particles. Twenty wild-type (WT) female C57BL/6j mice and 20 sclerostin-/- female C57BL/6j mice were used in this study. Mice were randomly divided into four groups: WT control group, WT SR group, knockout (KO) control group, and KO SR group. We found that SR enhanced the secretion of osteocalcin (0.72±0.007 in WT control group, 0.98±0.010 in WT SR group, P=0.000), Runx2 (0.34±0.005 in WT control group, 0.47±0.010 in WT SR group, P=0.000), β-catenin (1.04±0.05 in WT control group, 1.22±0.02 in WT SR group, P=0.000), and osteoprotegerin (OPG) (0.59±0.03 in WT control group, 0.90±0.02 in WT SR group, P=0.000). SR significantly decreased the level of receptor activator for nuclear factor-κB ligand (RANKL) (1.78±0.08 in WT control group, 1.37±0.06 in WT SR group, P=0.000) and improved the protein ratio of OPG/RANKL, but these effects were not observed in sclerostin-/- mice. Our findings demonstrated that SR enhanced bone formation and inhibited bone resorption in a wear particle-mediated osteolysis model in wild-type mice, and this effect relied mainly on the down-regulation of sclerostin levels to ameliorate the inhibition of the canonical Wnt pathway.
Collapse
Affiliation(s)
- Tianxiang Geng
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shouxuan Sun
- Department of Orthopedic Surgery, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Haochen Yu
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Haohui Guo
- Department of Orthopedic Surgery, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Mengxue Zheng
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shuai Zhang
- Department of Orthopedic Surgery, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xi Chen
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Qunhua Jin
- Department of Orthopedic Surgery, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Numerous forms of osteoporosis in childhood are characterized by low bone turnover (for example, osteoporosis due to neuromuscular disorders and glucocorticoid exposure). Anti-resorptive therapy, traditionally used to treat osteoporosis in the young, is associated with further reductions in bone turnover, raising concerns about the long-term safety and efficacy of such therapy. These observations have led to increasing interest in the role of anabolic therapy to treat pediatric osteoporosis. RECENT FINDINGS While growth hormone and androgens appears to be relatively weak anabolic modulators of bone mass, emerging therapies targeting bone formation pathways (anti-transforming growth factor beta antibody and anti-sclerostin antibody) hold considerable promise. Teriparatide remains an attractive option that merits formal study for patients post-epiphyseal fusion, although it must be considered that adult studies have shown its effect is blunted when administered following bisphosphonate therapy. Mechanical stimulation of bone through whole body vibration therapy appears to be much less effective than bisphosphonate therapy for treating osteoporosis in children. New anabolic therapies which target important pathways in skeletal metabolism merit further study in children, including their effects on fracture risk reduction and after treatment discontinuation.
Collapse
Affiliation(s)
- Leanne M Ward
- Department of Pediatrics, Faculty of Medicine, University of Ottawa and Division of Endocrinology and Metabolism, Children's Hospital of Eastern Ontario, 401 Smyth Road, Ottawa, Ontario, K1H 8L1, Canada.
| | - Frank Rauch
- Department of Pediatrics, Faculty of Medicine, McGill University, and Shriners Hospital for Children, 1003 Boulevard Décarie, Montréal, Québec, H4A 0A9, Canada
| |
Collapse
|
28
|
Bernardes M, Vieira T, Lucas R, Pereira J, Costa L, Simões-Ventura F, Martins MJ. Serum serotonin levels and bone in rheumatoid arthritis patients. Rheumatol Int 2017; 37:1891-1898. [PMID: 28993870 DOI: 10.1007/s00296-017-3836-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/23/2017] [Indexed: 10/18/2022]
Abstract
In rheumatoid arthritis (RA), a disease characterized by bone loss, increased levels of serotonin have been reported. Recent studies have demonstrated a role for circulating serotonin as a regulator of osteoblastogenesis, inhibiting bone formation. Thus, we measured serum serotonin levels (SSL) in a Portuguese sample of 205 RA patients and related these to anthropometric variables, disease parameters, serum bone biomarkers, and bone mineral density (BMD) assessed by dual-energy X-ray absorptiometry at several sites (total proximal femur, lumbar spine, left hand, and left second proximal phalange). SSL were inversely associated with body mass index (BMI) in RA women (r = - 0.218; p = 0.005), independent of exposure to biologics and/or bisphosphonates. Among biologic naïves, there was an inverse association between SSL and osteoprotegerin in RA women (r = - 0.260; p = 0.022). Serum β-CTX and dickkopf-1 were strongly associated with SSL in RA men not treated with bisphosphonates (r = 0.590; p < 0.001/r = 0.387; p = 0.031, respectively). There was also an inverse association between SSL and sclerostin in RA men (r = - 0.374; p < 0.05), stronger among biologic naïve or bisphosphonates-unexposed RA men. In crude models, SSL presented as a significant negative predictor of total proximal femur BMD in RA women as well as in postmenopausal RA women. After adjustment for BMI, disease duration, and years of menopause, SSL remained a significant negative predictor of total proximal femur BMD only in postmenopausal RA women. Our data reinforce a role, despite weak, for circulating serotonin in regulating bone mass in RA patients, with some differences in terms of gender and anatomical sites.
Collapse
Affiliation(s)
- Miguel Bernardes
- Department of Rheumatology, São João Hospital Center, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal. .,Medicine Department, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Tiago Vieira
- Department of Nuclear Medicine, São João Hospital Center, Porto, Portugal
| | - Raquel Lucas
- EPIUnit-Institute of Public Health, University of Porto, Porto, Portugal.,Department of Clinical Epidemiology, Predictive Medicine and Public Health, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Jorge Pereira
- Department of Nuclear Medicine, São João Hospital Center, Porto, Portugal
| | - Lúcia Costa
- Department of Rheumatology, São João Hospital Center, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | | | - Maria João Martins
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3s), University of Porto, Porto, Portugal
| |
Collapse
|
29
|
Abstract
A role for low-density lipoprotein-related receptor 5 (LRP5) in human bone was first established by the identification of genetic alterations that led to dramatic changes in bone mass. Shortly thereafter, mutations that altered the function of the sclerostin (SOST) gene were also associated with altered human bone mass. Subsequent studies of LRP5 and sclerostin have provided important insights into the mechanisms by which these proteins regulate skeletal homeostasis. Sclerostin normally binds to LRP5 and the related LRP6 protein and prevents their activation by Wnts, the LRP5/LRP6 ligands. The interaction of sclerostin with LRP5 or LRP6 is facilitated by the LRP4 protein. Loss of LRP5 leads to defective osteoblast function and low bone mass, while loss of SOST or mutations in LRP5, which produce a protein that can no longer be bound by SOST, result in high bone mass. Insights gained from the use of genetically engineered mouse models are presented, as well as a brief summary of the status of antibodies in clinical trials that block the function of SOST as a mechanism to increase bone mass.
Collapse
Affiliation(s)
- Bart O Williams
- Center for Cancer and Cell Biology and Program for Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute, United States.
| |
Collapse
|
30
|
Huang JY, Guo D. [SOST knockdown promotes differentiation of osteoblasts MG63 and mesenchymal stem cells C3H10 in an in vitro model of bone metastasis of breast cancer]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1035-1039. [PMID: 28801282 PMCID: PMC6765733 DOI: 10.3969/j.issn.1673-4254.2017.08.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To investigate whether SOST is involved in breast cancer MDA-MB-231 cells-induced suppression of differentiation of osteoblast MG63 cells and mesenchymal stem C3H10 cells. METHODS SOST-specific small interfering RNA (siRNA) was transfected into breast cancer MDA-MB-231 cells, and the interfering efficiency was verified by RT-PCR. The supernatants were collected from MDA-MB-231 cells in routine culture, cells transfected with SOST siRNA via adenovirus, and cells transfected with empty adenoviral vectors and added in MG63 or C3H10 cell cultures. The changes in the expressions of OPG, OCN, OPN and IBSP in MG63 and C3H10 cells were detected using quantitative real-time PCR, and ALP activity was detected with ALP reading and ALP staining with the cells cultured in routine culture medium and cells in osteogenic induction medium as the negative and positive controls. RESULTS The adenovirus Ad-siSOST effectively knocked down the expression of SOST in MDA-MB-231 cells. MG63 cells and C3H10 cells cultured in osteogenic medium showed significantly upregulated expressions of the osteoblast markers OPG, OPN, OCN and IBSP (P<0.01), while co-culture with the supernatant of MDA-MB-231 cells obviously reduced the expressions of the osteoblast markers (P<0.01); the expression of the markers increased again in MG63 and C3H10 cells after treatment with the supernatant of MDA-MB-231 cells transfected with ad-siSOST (P<0.01). ALP activity in MG63 and C3H10 cells exhibited a similar pattern of variations in response to the treatments (P<0.01). CONCLUSION In the in vitro model of bone metastasis of breast cancer, the differentiation of MG63 or C3H10 cells is suppressed, which can be partly reversed by knocking down the expression of SOST in the bone metastasis microenvironment.
Collapse
Affiliation(s)
- Jia-Yi Huang
- 1Department of Pathophysiology, 2Research Center of Molecular Medicine and Cancer, Chongqing Medical University, Chongqing 400016, China.E-mail:
| | | |
Collapse
|
31
|
Little DG, Peacock L, Mikulec K, Kneissel M, Kramer I, Cheng TL, Schindeler A, Munns C. Combination sclerostin antibody and zoledronic acid treatment outperforms either treatment alone in a mouse model of osteogenesis imperfecta. Bone 2017; 101:96-103. [PMID: 28461254 DOI: 10.1016/j.bone.2017.04.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 10/19/2022]
Abstract
In this study, we examined the therapeutic potential of anti-Sclerostin Antibody (Scl-Ab) and bisphosphonate treatments for the bone fragility disorder Osteogenesis Imperfecta (OI). Mice with the Amish OI mutation (Col1a2 G610C mice) and control wild type littermates (WT) were treated from week 5 to week 9 of life with (1) saline (control), (2) zoledronic acid given 0.025mg/kg s.c. weekly (ZA), (3) Scl-Ab given 50mg/kg IV weekly (Scl-Ab), or (4) a combination of both (Scl-Ab/ZA). Functional outcomes were prioritized and included bone mineral density (BMD), bone microarchitecture, long bone bending strength, and vertebral compression strength. By dual-energy absorptiometry, Scl-Ab treatment alone had no effect on tibial BMD, while ZA and Scl-Ab/ZA significantly enhanced BMD by week 4 (+16% and +27% respectively, P<0.05). Scl-Ab/ZA treatment also led to increases in cortical thickness and tissue mineral density, and restored the tibial 4-point bending strength to that of control WT mice. In the spine, all treatments increased compression strength over controls, but only the combined group reached the strength of WT controls. Scl-Ab showed greater anabolic effects in the trabecular bone than in cortical bone. In summary, the Scl-Ab/ZA intervention was superior to either treatment alone in this OI mouse model, however further studies are required to establish its efficacy in other preclinical and clinical scenarios.
Collapse
Affiliation(s)
- David G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, NSW, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia.
| | - Lauren Peacock
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Kathy Mikulec
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Michaela Kneissel
- Bone Unit, Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Ina Kramer
- Bone Unit, Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Tegan L Cheng
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, NSW, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, NSW, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Craig Munns
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, NSW, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
32
|
Ahn Y, Sims C, Murray MJ, Kuhlmann PK, Fuentes-Antrás J, Weatherbee SD, Krumlauf R. Multiple modes of Lrp4 function in modulation of Wnt/β-catenin signaling during tooth development. Development 2017; 144:2824-2836. [PMID: 28694256 DOI: 10.1242/dev.150680] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/30/2017] [Indexed: 12/29/2022]
Abstract
During development and homeostasis, precise control of Wnt/β-catenin signaling is in part achieved by secreted and membrane proteins that negatively control activity of the Wnt co-receptors Lrp5 and Lrp6. Lrp4 is related to Lrp5/6 and is implicated in modulation of Wnt/β-catenin signaling, presumably through its ability to bind to the Wise (Sostdc1)/sclerostin (Sost) family of Wnt antagonists. To gain insights into the molecular mechanisms of Lrp4 function in modulating Wnt signaling, we performed an array of genetic analyses in murine tooth development, where Lrp4 and Wise play important roles. We provide genetic evidence that Lrp4 mediates the Wnt inhibitory function of Wise and also modulates Wnt/β-catenin signaling independently of Wise. Chimeric receptor analyses raise the possibility that the Lrp4 extracellular domain interacts with Wnt ligands, as well as the Wnt antagonists. Diverse modes of Lrp4 function are supported by severe tooth phenotypes of mice carrying a human mutation known to abolish Lrp4 binding to Sost. Our data suggest a model whereby Lrp4 modulates Wnt/β-catenin signaling via interaction with Wnt ligands and antagonists in a context-dependent manner.
Collapse
Affiliation(s)
- Youngwook Ahn
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA .,Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Carrie Sims
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Megan J Murray
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Paige K Kuhlmann
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | - Scott D Weatherbee
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Robb Krumlauf
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA .,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
33
|
Ma YL, Hamang M, Lucchesi J, Bivi N, Zeng Q, Adrian MD, Raines SE, Li J, Kuhstoss SA, Obungu V, Bryant HU, Krishnan V. Time course of disassociation of bone formation signals with bone mass and bone strength in sclerostin antibody treated ovariectomized rats. Bone 2017; 97:20-28. [PMID: 27939957 DOI: 10.1016/j.bone.2016.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/02/2016] [Accepted: 12/07/2016] [Indexed: 10/20/2022]
Abstract
Sclerostin antibodies increase bone mass by stimulating bone formation. However, human and animal studies show that bone formation increases transiently and returns to pre-treatment level despite ongoing antibody treatment. To understand its mechanism of action, we studied the time course of bone formation, correlating the rate and extent of accrual of bone mass and strength after sclerostin antibody treatment. Ovariectomized (OVX) rats were treated with a sclerostin-antibody (Scle-ab) at 20mg/kg sc once weekly and sacrificed at baseline and 2, 3, 4, 6, and 8weeks post-treatment. In Scle-ab treated rats, serum PINP and OCN rapidly increased at week 1, peaked around week 3, and returned to OVX control levels by week 6. Transcript analyses from the distal femur revealed an early increase in bone formation followed by a sustained decrease in bone resorption genes. Lumbar vertebral (LV) osteoblast surface increased 88% by week 2, and bone formation rate (BFR/BS) increased 138% by week 4. Both parameters were below OVX control by week 8. Bone formation was primarily a result of modeling based formation. Endocortical and periosteal BFR/BS peaked around week 4 at 313% and 585% of OVX control, respectively. BFR/BS then declined but remained higher than OVX control on both surfaces through week 8. Histomorphometric analyses showed LV-BV/TV did not further increase after week 4, while BMD continued to increase at LV, mid femur (MF), and femoral neck (FN) through week 8. Biomechanical tests showed a similar improvement in bone strength through 8weeks in MF and FN, but bone strength plateaued between weeks 6 and 8 for LV. Our data suggest that bone formation with Scle-ab treatment is rapid and modeling formation dominated in OVX rats. Although transient, the bone formation response persists longer in cortical than trabecular bone.
Collapse
Affiliation(s)
- Yanfei L Ma
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA.
| | - Matthew Hamang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jonathan Lucchesi
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Nicoletta Bivi
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Qianqiang Zeng
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mary D Adrian
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Sarah E Raines
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jiliang Li
- Indiana University-Purdue University, Indianapolis, IN, USA
| | - Stuart A Kuhstoss
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Victor Obungu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Henry U Bryant
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Venkatesh Krishnan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
34
|
Screening for Key Pathways Associated with the Development of Osteoporosis by Bioinformatics Analysis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8589347. [PMID: 28466021 PMCID: PMC5390640 DOI: 10.1155/2017/8589347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/13/2017] [Accepted: 01/26/2017] [Indexed: 01/12/2023]
Abstract
Objectives. We aimed to find the key pathways associated with the development of osteoporosis. Methods. We downloaded expression profile data of GSE35959 and analyzed the differentially expressed genes (DEGs) in 3 comparison groups (old_op versus middle, old_op versus old, and old_op versus senescent). KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichment analyses were carried out. Besides, Venn diagram analysis and gene functional interaction (FI) network analysis were performed. Results. Totally 520 DEGs, 966 DEGs, and 709 DEGs were obtained in old_op versus middle, old_op versus old, and old_op versus senescent groups, respectively. Lysosome pathway was the significantly enriched pathways enriched by intersection genes. The pathways enriched by subnetwork modules suggested that mitotic metaphase and anaphase and signaling by Rho GTPases in module 1 had more proteins from module. Conclusions. Lysosome pathway, mitotic metaphase and anaphase, and signaling by Rho GTPases may be involved in the development of osteoporosis. Furthermore, Rho GTPases may regulate the balance of bone resorption and bone formation via controlling osteoclast and osteoblast. These 3 pathways may be regarded as the treatment targets for osteoporosis.
Collapse
|
35
|
Jacobsen CM. Application of anti-Sclerostin therapy in non-osteoporosis disease models. Bone 2017; 96:18-23. [PMID: 27780792 PMCID: PMC5328800 DOI: 10.1016/j.bone.2016.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/17/2016] [Accepted: 10/20/2016] [Indexed: 12/29/2022]
Abstract
Sclerostin, a known inhibitor of the low density lipoprotein related protein 5 and 6 (LRP5 and LRP6) cell surface signaling receptors, is integral in the maintenance of normal bone mass and strength. Patients with loss of function mutations in SOST or missense mutations in LRP5 that prevent Sclerostin from binding and inhibiting the receptor, have significantly increased bone mass. This observation leads to the development of Sclerostin neutralizing therapies to increase bone mass and strength. Anti-Sclerostin therapy has been shown to be effective at increasing bone density and strength in animal models and patients with osteoporosis. Loss of function of Sost or treatment with a Sclerostin neutralizing antibody improves bone properties in animal models of Osteoporosis Pseudoglioma syndrome (OPPG), likely due to action through the LRP6 receptor, which suggests patients may benefit from these therapies. Sclerostin antibody is effective at improving bone properties in mouse models of Osteogenesis Imperfecta, a genetic disorder of low bone mass and fragility due to type I collagen mutations, in as little as two weeks after initiation of therapy. However, these improvements are due to increases in bone quantity as the quality (brittleness) of bone remains unaffected. Similarly, Sclerostin antibody treatment improves bone density in animal models of other diseases. Sclerostin neutralizing therapies are likely to benefit many patients with genetic disorders of bone, as well as other forms of metabolic bone disease.
Collapse
Affiliation(s)
- Christina M Jacobsen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Boston Children's Hospital, Boston, MA, United States; Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States; Division of Genetics, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
36
|
Kang KS, Hong JM, Robling AG. Postnatal β-catenin deletion from Dmp1-expressing osteocytes/osteoblasts reduces structural adaptation to loading, but not periosteal load-induced bone formation. Bone 2016; 88:138-145. [PMID: 27143110 PMCID: PMC4899196 DOI: 10.1016/j.bone.2016.04.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 04/19/2016] [Accepted: 04/25/2016] [Indexed: 01/20/2023]
Abstract
Mechanical signal transduction in bone tissue begins with load-induced activation of several cellular pathways in the osteocyte population. A key pathway that participates in mechanotransduction is Wnt/Lrp5 signaling. A putative downstream mediator of activated Lrp5 is the nucleocytoplasmic shuttling protein β-catenin (βcat), which migrates to the nucleus where it functions as a transcriptional co-activator. We investigated whether osteocytic βcat participates in Wnt/Lrp5-mediated mechanotransduction by conducting ulnar loading experiments in mice with or without chemically induced βcat deletion in osteocytes. Mice harboring βcat floxed loss-of-function alleles (βcat(f/f)) were bred to the inducible osteocyte Cre transgenic (10)(kb)Dmp1-CreERt2. Adult male mice were induced to recombine the βcat alleles using tamoxifen, and intermittent ulnar loading sessions were applied over the following week. Although adult-onset deletion of βcat from Dmp1-expressing cells reduced skeletal mass, the bone tissue was responsive to mechanical stimulation as indicated by increased relative periosteal bone formation rates in recombined mice. However, load-induced improvements in cross sectional geometric properties were compromised in recombined mice. The collective results indicate that the osteoanabolic response to loading can occur on the periosteal surface when β-cat levels are significantly reduced in Dmp1-expressing cells, suggesting that either (i) only low levels of β-cat are required for mechanically induced bone formation on the periosteal surface, or (ii) other additional downstream mediators of Lrp5 might participate in transducing load-induced Wnt signaling.
Collapse
Affiliation(s)
- Kyung Shin Kang
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jung Min Hong
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander G Robling
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA; Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
37
|
Neben CL, Roberts RR, Dipple KM, Merrill AE, Klein OD. Modeling craniofacial and skeletal congenital birth defects to advance therapies. Hum Mol Genet 2016; 25:R86-R93. [PMID: 27346519 DOI: 10.1093/hmg/ddw171] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/24/2016] [Indexed: 12/12/2022] Open
Abstract
Craniofacial development is an intricate process of patterning, morphogenesis, and growth that involves many tissues within the developing embryo. Genetic misregulation of these processes leads to craniofacial malformations, which comprise over one-third of all congenital birth defects. Significant advances have been made in the clinical management of craniofacial disorders, but currently very few treatments specifically target the underlying molecular causes. Here, we review recent studies in which modeling of craniofacial disorders in primary patient cells, patient-derived induced pluripotent stem cells (iPSCs), and mice have enhanced our understanding of the etiology and pathophysiology of these disorders while also advancing therapeutic avenues for their prevention.
Collapse
Affiliation(s)
- Cynthia L Neben
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Ryan R Roberts
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Katrina M Dipple
- Departments of Pediatrics and Human Genetics, David Geffen School of Medicine and InterDepartmental Program Biomedical Engineering, Henry Samulei School of Engineering and Applied Sciences, University of California, Los Angeles, CA, USA
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
38
|
Chapurlat R. In vivo evaluation of bone microstructure in humans: Clinically useful? BONEKEY REPORTS 2016; 5:813. [PMID: 27347398 DOI: 10.1038/bonekey.2016.40] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/13/2016] [Accepted: 02/22/2016] [Indexed: 11/09/2022]
Abstract
In vivo evaluation of bone microstructure with high-resolution peripheral quantitative tomography (HRpQCT) has been used for a decade in research settings. In this review, we examine the value this technique could have in clinical practice. Bone microstructure parameters obtained with HRpQCT are associated with prevalent fracture in men and women. In postmenopausal women, some parameters also predict incident fracture, independently of areal bone mineral density. In specific population groups including patients with diabetes, chronic kidney disease, glucocorticosteroid therapy and rheumatic diseases, abnormal microstructure parameters from HRpQCT have been reported. Findings from HRpQCT studies may also explain ethnic differences in bone fragility. Treatment monitoring has been challenging in the various clinical trials with available HRpQCT data. The improvements were of small magnitude but tended to be proportional to the potency of antiresorptive agents. Microfinite element analysis was a better predictor of treatment efficacy than the microarchitectural parameters. In conclusion, HRpQCT remains a valuable research tool, but more work is needed to be able to use it in clinical practice.
Collapse
Affiliation(s)
- Roland Chapurlat
- INSERM UMR 1033, Université de Lyon, Hôpital E Herriot , Lyon, France
| |
Collapse
|
39
|
MacNabb C, Patton D, Hayes JS. Sclerostin Antibody Therapy for the Treatment of Osteoporosis: Clinical Prospects and Challenges. J Osteoporos 2016; 2016:6217286. [PMID: 27313945 PMCID: PMC4899597 DOI: 10.1155/2016/6217286] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/21/2016] [Indexed: 01/22/2023] Open
Abstract
It is estimated that over 200 million adults worldwide have osteoporosis, a disease that has increasing socioeconomic impact reflected by unsustainable costs associated with disability, fracture management, hospital stays, and treatment. Existing therapeutic treatments for osteoporosis are associated with a variety of issues relating to use, clinical predictability, and health risks. Consequently, additional novel therapeutic targets are increasingly sought. A promising therapeutic candidate is sclerostin, a Wnt pathway antagonist and, as such, a negative regulator of bone formation. Sclerostin antibody treatment has demonstrated efficacy and superiority compared to other anabolic treatments for increasing bone formation in both preclinical and clinical settings. Accordingly, it has been suggested that sclerostin antibody treatment is set to achieve market approval by 2017 and aggressively compete as the gold standard for osteoporotic treatment by 2021. In anticipation of phase III trial results which may potentially signify a significant step in achieving market approval here, we review the preclinical and clinical emergence of sclerostin antibody therapies for both osteoporosis and alternative applications. Potential clinical challenges are also explored as well as ongoing developments that may impact on the eventual clinical application of sclerostin antibodies as an effective treatment of osteoporosis.
Collapse
Affiliation(s)
- Claire MacNabb
- Regenerative Medicine Institute, NUI Galway, Biosciences Research Building, Corrib Village, Dangan, Galway, Ireland
| | - D. Patton
- Regenerative Medicine Institute, NUI Galway, Biosciences Research Building, Corrib Village, Dangan, Galway, Ireland
| | - J. S. Hayes
- Regenerative Medicine Institute, NUI Galway, Biosciences Research Building, Corrib Village, Dangan, Galway, Ireland
| |
Collapse
|
40
|
Erjavec I, Bordukalo-Niksic T, Brkljacic J, Grcevic D, Mokrovic G, Kesic M, Rogic D, Zavadoski W, Paralkar VM, Grgurevic L, Trkulja V, Cicin-Sain L, Vukicevic S. Constitutively Elevated Blood Serotonin Is Associated with Bone Loss and Type 2 Diabetes in Rats. PLoS One 2016; 11:e0150102. [PMID: 26907598 PMCID: PMC4764355 DOI: 10.1371/journal.pone.0150102] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 02/09/2016] [Indexed: 11/19/2022] Open
Abstract
Reduced peripheral serotonin (5HT) in mice lacking tryptophan hydroxylase (TPH1), the rate limiting enzyme for 5HT synthesis, was reported to be anabolic to the skeleton. However, in other studies TPH1 deletion either had no bone effect or an age dependent inhibition of osteoclastic bone resorption. The role of 5HT in bone therefore remains poorly understood. To address this issue, we used selective breeding to create rat sublines with constitutively high (high-5HT) and low (low-5HT) platelet 5HT level (PSL) and platelet 5HT uptake (PSU). High-5HT rats had decreased bone volume due to increased bone turnover characterized by increased bone formation and mineral apposition rate, increased osteoclast number and serum C-telopeptide level. Daily oral administration of the TPH1 inhibitor (LX1032) for 6 weeks reduced PSL and increased the trabecular bone volume and trabecular number of the spine and femur in high-5HT rats. High-5HT animals also developed a type 2 diabetes (T2D) phenotype with increased: plasma insulin, glucose, hemoglobin A1c, body weight, visceral fat, β-cell pancreatic islets size, serum cholesterol, and decreased muscle strength. Serum calcium accretion mediated by parathyroid hormone slightly increased, whereas treatment with 1,25(OH)2D3 decreased PSL. Insulin reduction was paralleled by a drop in PSL in high-5HT rats. In vitro, insulin and 5HT synergistically up-regulated osteoblast differentiation isolated from high-5HT rats, whereas TPH1 inhibition decreased the number of bone marrow-derived osteoclasts. These results suggest that constitutively elevated PSL is associated with bone loss and T2D via a homeostatic interplay between the peripheral 5HT, bone and insulin.
Collapse
Affiliation(s)
- Igor Erjavec
- Laboratory of Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Tatjana Bordukalo-Niksic
- Laboratory of Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Jelena Brkljacic
- Laboratory of Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Danka Grcevic
- Department of Physiology and Immunology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Gordana Mokrovic
- Laboratory for Neurochemistry and Molecular Neurobiology, Molecular Biology Department, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Maja Kesic
- Laboratory for Neurochemistry and Molecular Neurobiology, Molecular Biology Department, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Dunja Rogic
- Clinical Hospital Center Zagreb, Zagreb, Croatia
| | | | | | - Lovorka Grgurevic
- Laboratory of Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Vladimir Trkulja
- Department of Pharmacology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Lipa Cicin-Sain
- Laboratory for Neurochemistry and Molecular Neurobiology, Molecular Biology Department, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Slobodan Vukicevic
- Laboratory of Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
41
|
Thouverey C, Caverzasio J. Sclerostin inhibits osteoblast differentiation without affecting BMP2/SMAD1/5 or Wnt3a/β-catenin signaling but through activation of platelet-derived growth factor receptor signaling in vitro. BONEKEY REPORTS 2015; 4:757. [PMID: 26587226 DOI: 10.1038/bonekey.2015.126] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/29/2015] [Indexed: 01/18/2023]
Abstract
Sclerostin inhibits bone formation mostly by antagonizing LRP5/6, thus inhibiting Wnt signaling. However, experiments with genetically modified mouse models suggest that a significant part of sclerostin-mediated inhibition of bone formation is due to interactions with other binding partners. The objective of the present work was to identify signaling pathways affected by sclerostin in relation with its inhibitory action on osteogenic differentiation of C3H10T1/2 cells, MC3T3-E1 cells and primary osteoblasts. Sclerostin inhibited BMP2-induced osteoblast differentiation without altering SMAD1/5 phosphorylation and transcriptional activity. Moreover, sclerostin prevented Wnt3a-mediated osteoblastogenesis without affecting LRP5/6 phosphorylation or β-catenin transcriptional activity. In addition, sclerostin inhibited mineralization promoted by GSK3 inhibition, which mimics canonical Wnt signaling without activation of LRP5/6, suggesting that sclerostin can prevent osteoblast differentiation without antagonizing LRP5/6. Finally, we found that sclerostin could activate platelet-derived growth factor receptor (PDGFR) and its downstream signaling pathways PLCγ, PKC, Akt and ERK1/2. PDGFR inhibition could reverse sclerostin-mediated inhibitory activity on BMP2-induced osteoblast differentiation. Therefore, our data suggest that sclerostin can activate PDGFR signaling by itself, and this functional interaction may be involved in the negative effect of sclerostin on osteoblast differentiation.
Collapse
Affiliation(s)
- Cyril Thouverey
- Service of Bone Diseases, Department of Internal Medicine Specialties, University Hospital of Geneva , Geneva, Switzerland
| | - Joseph Caverzasio
- Service of Bone Diseases, Department of Internal Medicine Specialties, University Hospital of Geneva , Geneva, Switzerland
| |
Collapse
|
42
|
Brommage R. Genetic Approaches To Identifying Novel Osteoporosis Drug Targets. J Cell Biochem 2015; 116:2139-45. [DOI: 10.1002/jcb.25179] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 03/30/2015] [Indexed: 12/26/2022]
|
43
|
Yorgan TA, Peters S, Jeschke A, Benisch P, Jakob F, Amling M, Schinke T. The Anti-Osteoanabolic Function of Sclerostin Is Blunted in Mice Carrying a High Bone Mass Mutation of Lrp5. J Bone Miner Res 2015; 30:1175-83. [PMID: 25640331 DOI: 10.1002/jbmr.2461] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 12/23/2014] [Accepted: 01/13/2015] [Indexed: 12/17/2022]
Abstract
Activating mutations of the putative Wnt co-receptor Lrp5 or inactivating mutations of the secreted molecule Sclerostin cause excessive bone formation in mice and humans. Previous studies have suggested that Sclerostin functions as an Lrp5 antagonist, yet clear in vivo evidence was still missing, and alternative mechanisms have been discussed. Moreover, because osteoblast-specific inactivation of β-catenin, the major intracellular mediator of canonical Wnt signaling, primarily affected bone resorption, it remained questionable, whether Sclerostin truly acts as a Wnt signaling antagonist by interacting with Lrp5. In an attempt to address this relevant question, we generated a mouse model (Col1a1-Sost) with transgenic overexpression of Sclerostin under the control of a 2.3-kb Col1a1 promoter fragment. These mice displayed the expected low bone mass phenotype as a consequence of reduced bone formation. The Col1a1-Sost mice were then crossed with two mouse lines carrying different high bone mass mutations of Lrp5 (Lrp5(A170V) and Lrp5(G213V)), both of them potentially interfering with Sclerostin binding. Using µCT-scanning and histomorphometry we found that the anti-osteoanabolic influence of Sclerostin overexpression was not observed in Lrp5(A213V/A213V) mice and strongly reduced in Lrp5(A170V/A170V) mice. As a control we applied the same strategy with mice overexpressing the transmembrane Wnt signaling antagonist Krm2 and found that the anti-osteoanabolic influence of the Col1a1-Krm2 transgene was not affected by either of the Lrp5 mutations. Taken together, our data support the concept that Sclerostin inhibits bone formation through Lrp5 interaction, yet their physiological relevance remains to be established.
Collapse
Affiliation(s)
- Timur A Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephanie Peters
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Jeschke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peggy Benisch
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
44
|
Tosi LL, Warman ML. Mechanistic and therapeutic insights gained from studying rare skeletal diseases. Bone 2015; 76:67-75. [PMID: 25819040 DOI: 10.1016/j.bone.2015.03.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 03/19/2015] [Indexed: 12/14/2022]
Abstract
Rare bone diseases account for 5% of all birth defects and can cause significant morbidity throughout patients' lives. Significant progress is being made to elucidate the pathophysiological mechanisms underlying these diseases. This paper summarizes presentation highlights of a workshop on Rare Skeletal Diseases convened to explore how the study of rare diseases has influenced the field's understanding of bone anabolism and catabolism and directed the search for new therapies benefiting patients with rare conditions as well as patients with common skeletal disorders.
Collapse
Affiliation(s)
- Laura L Tosi
- Division of Orthopaedics and Sports Medicine, Children's National Health System, 111 Michigan Avenue NW, Washington, DC 20010, USA.
| | - Matthew L Warman
- Orthopaedic Research Laboratories, Boston Children's Hospital, 320 Longwood Avenue, Room EN260.1, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Abstract
A dozen years ago the identification of causal mutations in the low-density lipoprotein receptor-related protein 5 (LRP5) gene involved in two rare bone disorders propelled research in the bone field in totally new directions. Since then, there have been an explosion in the number of reports that highlight the role of the Wnt/β-catenin pathway in the regulation of bone homeostasis. In this review we discuss some of the most recent reports (in the past 2 years) highlighting the involvement of the members of the LRP family (LRP5, LRP6, LRP4, and more recently LRP8) in the maintenance of bone and their implications in bone diseases. These reports include records of new single nucleotides polymorphisms (SNPs) and haplotypes that suggest variants in these genes can contribute to subtle variation in bone traits to mutations that give rise to extreme bone phenotypes. All of these serve to further support and reinforce the importance of this tightly regulated pathway in bone. Furthermore, we discuss provocative reports suggesting novel approaches through inhibitors of this pathway to treat rarer diseases such as Osteoporosis-Pseudoglioma Syndrome (OPPG), Osteogenesis Imperfecta (OI), and Sclerosteosis/Van Buchem disease. It is hoped that by understanding the role of each component of the pathway and their involvement in bone diseases that this knowledge will allow us to develop new, more effective therapeutic approaches for more common diseases such as post-menopausal osteoporosis, osteoarthritis, and rheumatoid arthritis as well as these rarer bone diseases.
Collapse
Affiliation(s)
- N Lara-Castillo
- Department of Oral and Craniofacial Sciences, UMKC School of Dentistry, 650 East 25th Street, Kansas City, MO, 64108, USA,
| | | |
Collapse
|
46
|
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: 351] [Impact Index Per Article: 35.1] [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
|
47
|
Alonso N, Soares DC, V McCloskey E, Summers GD, Ralston SH, Gregson CL. Atypical femoral fracture in osteoporosis pseudoglioma syndrome associated with two novel compound heterozygous mutations in LRP5. J Bone Miner Res 2015; 30:615-20. [PMID: 25384351 DOI: 10.1002/jbmr.2403] [Citation(s) in RCA: 20] [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: 08/15/2014] [Revised: 10/20/2014] [Accepted: 11/05/2014] [Indexed: 12/13/2022]
Abstract
Osteoporosis pseudoglioma syndrome (OPPG) is a rare autosomal recessive condition of congenital blindness and severe childhood osteoporosis with skeletal fragility, caused by loss-of-function mutations in the low-density lipoprotein receptor-related protein 5 (LRP5) gene. We report the first case of atypical (subtrochanteric) femoral fracture (AFF) in OPPG, occurring in a 38-year-old man within the context of relatively low bone turnover and trabecular osteoporosis on bone histology. We identify two novel LRP5 mutations: R752W is associated with low bone mineral density (BMD), as demonstrated by the heterozygous carriage identified in his 57-year-old mother; however, the combination of this R752W mutation with another novel W79R mutation, causes a severe case of compound heterozygous OPPG. We undertake 3D homology modeling of the four extracellular YWTD β-propeller/EGF-like domains (E1-E4) of LRP5, and show that both novel mutations destabilize the β-propeller domains that are critical for protein and ligand binding to regulate Wnt signaling and osteoblast function. Although AFFs have been reported in other rare bone diseases, this is the first in a genetic condition of primary osteoblast dysfunction. The relatively low bone turnover observed, and knowledge of LRP5 function, implicates impaired bone remodeling in the pathogenesis of AFF.
Collapse
Affiliation(s)
- Nerea Alonso
- Rheumatic Diseases Unit, Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | |
Collapse
|
48
|
Bouaziz W, Funck-Brentano T, Lin H, Marty C, Ea HK, Hay E, Cohen-Solal M. Loss of sclerostin promotes osteoarthritis in mice via β-catenin-dependent and -independent Wnt pathways. Arthritis Res Ther 2015; 17:24. [PMID: 25656376 PMCID: PMC4355467 DOI: 10.1186/s13075-015-0540-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 01/23/2015] [Indexed: 11/21/2022] Open
Abstract
Introduction Sclerostin is a Wnt inhibitor produced by osteocytes that regulates bone formation. Because bone tissue contributes to the development of osteoarthritis (OA), we investigated the role of sclerostin in bone and cartilage in a joint instability model in mice. Methods Ten-week-old SOST-knockout (SOST-KO) and wild-type (WT) mice underwent destabilization of the medial meniscus (DMM). We measured bone volume at the medial femoral condyle and osteophyte volume and determined the OA score and expression of matrix proteins. Primary murine chondrocytes were cultured with Wnt3a and sclerostin to assess the expression of matrix proteins, proteoglycan release and glycosaminoglycan accumulation. Results Sclerostin was expressed in calcified cartilage of WT mice with OA. In SOST-KO mice, cartilage was preserved despite high bone volume. However, SOST-KO mice with DMM had a high OA score, with increased expression of aggrecanases and type X collagen. Moreover, SOST-KO mice with OA showed disrupted anabolic–catabolic balance and cartilage damage. In primary chondrocytes, sclerostin addition abolished Wnt3a-increased expression of a disintegrin and metalloproteinase with thrombospondin motifs, matrix metalloproteinases and type X collagen by inhibiting the canonical Wnt pathway. Moreover, sclerostin inhibited Wnt-phosphorylated c-Jun N-terminal kinase (JNK) and rescued the expression of anabolic genes. Furthermore, sclerostin treatment inhibited both Wnt canonical and non-canonical JNK pathways in chondrocytes, thus preserving metabolism. Conclusion Sclerostin may play an important role in maintaining cartilage integrity in OA. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0540-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wafa Bouaziz
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Thomas Funck-Brentano
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Hilène Lin
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Caroline Marty
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Hang-Korng Ea
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Eric Hay
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| | - Martine Cohen-Solal
- Inserm U1132 and university Paris 7, hopital Lariboisiere, 2 rue Ambroise Pare, 75010, Paris, France.
| |
Collapse
|
49
|
Gerbaix M, Vico L, Ferrari SL, Bonnet N. Periostin expression contributes to cortical bone loss during unloading. Bone 2015; 71:94-100. [PMID: 25445447 DOI: 10.1016/j.bone.2014.10.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/19/2014] [Accepted: 10/15/2014] [Indexed: 01/26/2023]
Abstract
Periostin (a product of Postn gene) is a matricellular protein which is increased in periosteal osteoblasts and osteocytes upon mechanical stimulation. We previously reported that periostin-deficient mice (Postn(-/-)) have low bone mass and a diminished response to physical activity due to a lack of sclerostin (a product of Sost gene) inhibition by mechanical loading. Here we hypothesized that periostin could play a central role in the control of bone loss during unloading induced by hindlimb suspension (HU). In Postn(+/+) mice (wildtype littermate), HU significantly decreased femur BMD, as well as trabecular BV/TV and thickness (Tb.Th). Cortical bone volume and thickness at the femoral midshaft, also significantly decreased. These changes were explained by an inhibition of endocortical and periosteal bone formation activity and correlated with a decrease of Postn expression and a consecutive increase in Sost early after HU. Whereas trabecular bone loss in Postn(-/-) mice was comparable to Postn(+/+) mice, HU did not significantly alter cortical bone microstructure and strength in Postn(-/-) mice. Bone formation remained unchanged in these mice, as Sost did not increase in the absence of periostin. In contrast, changes in Dkk1, Rankl and Opg expression in response to HU were similar to Postn(+/+) mice, indicating that changes in periostin expression were quite specifically related to changes in Sost. In conclusion, HU inhibits periostin expression, which in turn plays an important role in cortical bone loss through an increase in Sost. These results further indicate that periostin is an essential mediator of cortical bone response to mechanical forces (loading and unloading).
Collapse
Affiliation(s)
- Maude Gerbaix
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospital & Faculty of Medicine, Geneva 14, Switzerland
| | - Laurence Vico
- Institute National de la Santé et de la Recherche Médicale (INSERM), Unité 1059, Laboratoire de Biologie Intégrée du Tissu Osseux, Université de Lyon, St-Etienne, France
| | - Serge L Ferrari
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospital & Faculty of Medicine, Geneva 14, Switzerland
| | - Nicolas Bonnet
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospital & Faculty of Medicine, Geneva 14, Switzerland.
| |
Collapse
|
50
|
Velázquez-Cruz R, Jiménez-Ortega RF, Parra-Torres AY, Castillejos-López M, Patiño N, Quiterio M, Villarreal-Molina T, Salmerón J. Analysis of association of MEF2C, SOST and JAG1 genes with bone mineral density in Mexican-Mestizo postmenopausal women. BMC Musculoskelet Disord 2014; 15:400. [PMID: 25430630 PMCID: PMC4258010 DOI: 10.1186/1471-2474-15-400] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/24/2014] [Indexed: 12/18/2022] Open
Abstract
Background Osteoporosis, a disease characterized by low bone mineral density (BMD), is an important health problem in Mexico. BMD is a highly heritable trait, with heritability estimates of 50-85%. Several candidate genes have been evaluated to identify those involved in BMD variation and the etiology of osteoporosis. This study investigated the possible association of single-nucleotide polymorphisms (SNPs) in the MEF2C, SOST and JAG1 genes with bone mineral density (BMD) variation in postmenopausal Mexican-Mestizo women. Methods Four hundred unrelated postmenopausal women were included in the study. Risk factors were recorded and BMD was measured in total hip, femoral neck and lumbar spine using dual-energy X-ray absorptiometry. In an initial stage, a total of twenty-five SNPs within or near SOST gene and seven SNPs in the JAG1 gene were genotyped using a GoldenGate assay. In a second stage, three MEF2C gene SNPs were also genotyped and SOST and JAG1 gene variants were validated. Real time PCR and TaqMan probes were used for genotyping. Results Linear regression analyses adjusted by age, body mass index and ancestry estimates, showed that five SNPs in the SOST gene were significantly associated with BMD in total hip and femoral neck but not lumbar spine. The lowest p value was 0.0012, well below the multiple–test significance threshold (p = 0.009), with mean effect size of -0.027 SD per risk allele. We did not find significant associations between BMD and MEF2C/JAG1 gene variants [rs1366594 “A” allele: β = 0.001 (95% CI -0.016; 0.017), P = 0.938; rs2273061 “G” allele: β = 0.007 (95% CI -0.007; 0.023), p = 0.409]. Conclusions SOST polymorphisms may contribute to total hip and femoral neck BMD variation in Mexican postmenopausal women. Together, these and prior findings suggest that this gene may contribute to BMD variation across populations of diverse ancestry. Electronic supplementary material The online version of this article (doi:10.1186/1471-2474-15-400) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica, Mexico City, Mexico.
| | | | | | | | | | | | | | | |
Collapse
|