1
|
Tsadaris SA, Komatsu DE, Grubisic V, Ramos RL, Hadjiargyrou M. A GCaMP reporter mouse with chondrocyte specific expression of a green fluorescent calcium indicator. Bone 2024; 188:117234. [PMID: 39147354 PMCID: PMC11392458 DOI: 10.1016/j.bone.2024.117234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
One of the major processes occurring during the healing of a fractured long bone is chondrogenesis, leading to the formation of the soft callus, which subsequently undergoes endochondral ossification and ultimately bridges the fracture site. Thus, understanding the molecular mechanisms of chondrogenesis can enhance our knowledge of the fracture repair process. One such molecular process is calciun (Ca++) signaling, which is known to play a critical role in the development and regeneration of multiple tissues, including bone, in response to external stimuli. Despite the existence of various mouse models for studying Ca++ signaling, none of them were designed to specifically examine the skeletal system or the various musculoskeletal cell types. As such, we generated a genetically engineered mouse model that is specific to cartilage (crossed with Col2a1 Cre mice) to study chondrocytes. Herein, we report on the characterization of this transgenic mouse line using conditional expression of GCaMP6f, a Ca++-indicator protein. Specifically, this mouse line exhibits increased GCaMP6f fluorescence following Ca++ binding in chondrocytes. Using this model, we show real-time Ca++ signaling in embryos, newborn and adult mice, as well as in fracture calluses. Further, robust expression of GCaMP6f in chondrocytes can be easily detected in embryos, neonates, adults, and fracture callus tissue sections. Finally, we also report on Ca++ signaling pathway gene expression, as well as real-time Ca++ transient measurements in fracture callus chondrocytes. Taken together, these mice provide a new experimental tool to study chondrocyte-specific Ca++ signaling during skeletal development and regeneration, as well as various in vitro perturbations.
Collapse
Affiliation(s)
- Sotirios A Tsadaris
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | - David E Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, Stony Brook, NY, USA
| | - Vladimir Grubisic
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, USA; Center for Biomedical Innovation, College of Osteopathic Medicine, New York Institute of Technology, USA
| | - Raddy L Ramos
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, USA
| | - Michael Hadjiargyrou
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA.
| |
Collapse
|
2
|
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
|
3
|
Chandran M, Akesson KE, Javaid MK, Harvey N, Blank RD, Brandi ML, Chevalley T, Cinelli P, Cooper C, Lems W, Lyritis GP, Makras P, Paccou J, Pierroz DD, Sosa M, Thomas T, Silverman S. Impact of osteoporosis and osteoporosis medications on fracture healing: a narrative review. Osteoporos Int 2024; 35:1337-1358. [PMID: 38587674 PMCID: PMC11282157 DOI: 10.1007/s00198-024-07059-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/06/2024] [Indexed: 04/09/2024]
Abstract
Antiresorptive medications do not negatively affect fracture healing in humans. Teriparatide may decrease time to fracture healing. Romosozumab has not shown a beneficial effect on human fracture healing. BACKGROUND Fracture healing is a complex process. Uncertainty exists over the influence of osteoporosis and the medications used to treat it on fracture healing. METHODS Narrative review authored by the members of the Fracture Working Group of the Committee of Scientific Advisors of the International Osteoporosis Foundation (IOF), on behalf of the IOF and the Société Internationale de Chirurgie Orthopédique et de Traumatologie (SICOT). RESULTS Fracture healing is a multistep process. Most fractures heal through a combination of intramembranous and endochondral ossification. Radiographic imaging is important for evaluating fracture healing and for detecting delayed or non-union. The presence of callus formation, bridging trabeculae, and a decrease in the size of the fracture line over time are indicative of healing. Imaging must be combined with clinical parameters and patient-reported outcomes. Animal data support a negative effect of osteoporosis on fracture healing; however, clinical data do not appear to corroborate with this. Evidence does not support a delay in the initiation of antiresorptive therapy following acute fragility fractures. There is no reason for suspension of osteoporosis medication at the time of fracture if the person is already on treatment. Teriparatide treatment may shorten fracture healing time at certain sites such as distal radius; however, it does not prevent non-union or influence union rate. The positive effect on fracture healing that romosozumab has demonstrated in animals has not been observed in humans. CONCLUSION Overall, there appears to be no deleterious effect of osteoporosis medications on fracture healing. The benefit of treating osteoporosis and the urgent necessity to mitigate imminent refracture risk after a fracture should be given prime consideration. It is imperative that new radiological and biological markers of fracture healing be identified. It is also important to synthesize clinical and basic science methodologies to assess fracture healing, so that a convergence of the two frameworks can be achieved.
Collapse
Affiliation(s)
- M Chandran
- Osteoporosis and Bone Metabolism Unit, Department of Endocrinology, Singapore General Hospital, DUKE NUS Medical School, Singapore, Singapore.
| | - K E Akesson
- Clinical and Molecular Osteoporosis Research Unit, Department of Clinical Sciences, Lund University, Department of Orthopedics, Skåne University Hospital, Malmö, Sweden
| | - M K Javaid
- NIHR Musculoskeletal Biomedical Research Unit, University of Oxford, Oxford, UK
| | - N Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - R D Blank
- Garvan Institute of Medical Research, Medical College of Wisconsin, Darlinghurst, NSW, Australia
- Medical College of Wisconsin, Milwaukee, WI, USA
| | - M L Brandi
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Largo Palagi 1, Florence, Italy
| | - T Chevalley
- Division of Bone Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - P Cinelli
- Department of Trauma Surgery, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - C Cooper
- MRC Lifecourse Epidemiology Centre, University of Southampton, NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospitals Southampton NHS Foundation Trust, Southampton, UK
- NIHR Oxford Biomedical Research Unit, University of Oxford, Oxford, UK
| | - W Lems
- Department of Rheumatology, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - G P Lyritis
- Hellenic Osteoporosis Foundation, Athens, Greece
| | - P Makras
- Department of Medical Research, 251 Hellenic Air Force & VA General Hospital, Athens, Greece
| | - J Paccou
- Department of Rheumatology, MABlab ULR 4490, CHU Lille, Univ. Lille, 59000, Lille, France
| | - D D Pierroz
- International Osteoporosis Foundation, Nyon, Switzerland
| | - M Sosa
- University of Las Palmas de Gran Canaria, Investigation Group on Osteoporosis and Mineral Metabolism, Canary Islands, Spain
| | - T Thomas
- Department of Rheumatology, North Hospital, CHU Saint-Etienne and INSERM U1059, University of Lyon-University Jean Monnet, Saint‑Etienne, France
| | - S Silverman
- Cedars-Sinai Medical Center and Geffen School of Medicine UCLA, Los Angeles, CA, USA
| |
Collapse
|
4
|
Khoswanto C, Dewi IK. The role of Wnt signaling on Tooth Extraction Wound Healing: Narrative review. Saudi Dent J 2024; 36:516-520. [PMID: 38690381 PMCID: PMC11056418 DOI: 10.1016/j.sdentj.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 05/02/2024] Open
Abstract
Compared to an incisional skin or mucosal wound, a tooth extraction wound results in far more soft tissue loss. A blood clot instantly fills the gap left by the extracted tooth. An embryonic type of bone forms during the healing of extraction wounds, and mature bone only later replaces it. Osteocytes in embryonic bone, also known as coarse fibrillar bone or immature bone, differ from those in adult bone in terms of number, size, and irregular arrangement. This immature bone is more radiolucent than mature bone due to the higher cell density and the smaller volume of calcified intercellular material. The Wnt gene family contains genes that encode secreted signaling proteins that have good promise for promoting bone regeneration. However, we still have a limited understanding the interplay of the molecular elements of the Wnt pathway in signal transduction, from ligand detection on the cell surface to transcription of target genes in the nucleus. We discuss the function of Wnt signaling molecules in this review, in tissue repair following tooth extraction and present recent results about these molecules. Conclusions: Wnt signaling activity helps to hasten bone regeneration while bone healing is slowed down by mutations in LRP5/6 or β-catenin.
Collapse
Affiliation(s)
- Christian Khoswanto
- Department of Oral Biology Faculty of Dentistry, Airlangga University Surabaya, Indonesia
| | | |
Collapse
|
5
|
Yu BF, Wang Z, Chen XX, Zeng Q, Dai CC, Wei J. Continuous dynamic identification of key genes and molecular signaling pathways of periosteum in guided bone self-generation in swine model. J Orthop Surg Res 2023; 18:53. [PMID: 36653843 PMCID: PMC9847205 DOI: 10.1186/s13018-023-03524-y] [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: 11/18/2022] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Guided bone self-generation with periosteum-preserved has successfully regenerated mandibular, temporomandibular and interphalangeal joint. The aim of this study was to investigate the dynamic changes of gene expression of periosteum which was involved in the guided bone self-generation. METHODS Rib defects of critical size were created in mature swine with periosteum-preserved. The periosteum was sutured into a sealed sheath that closed the bone defect. The periosteum of trauma and control sites were harvested at postoperative 9 time points, and total RNA was extracted. Microarray analysis was conducted to identify the differences in the transcriptome of different time points between two groups. RESULTS The differentially expressed genes (DEGs) between control and trauma group were different at postoperative different time points. The dynamic changes of the number of DEGs fluctuated a lot. There were 3 volatility peaks, and we chose 3 time points of DEG number peak (1 week, 5 weeks and 6 months) to study the functions of DEGs. Oxidoreductase activity, oxidation-reduction process and mitochondrion are the most enriched terms of Go analysis. The major signaling pathways of DEGs enrichment include oxidative phosphorylation, PI3K-Akt signaling pathway, osteoclast differentiation pathway and Wnt signaling. CONCLUSIONS The oxidoreductase reaction was activated during this bone regeneration process. The oxidative phosphorylation, PI3K-Akt signaling pathway, osteoclast differentiation pathway and Wnt signaling may play important roles in the guided bone self-generation with periosteum-preserved. This study can provide a reference for how to improve the application of this concept of bone regeneration.
Collapse
Affiliation(s)
- Bao-Fu Yu
- grid.412523.30000 0004 0386 9086Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011 China
| | - Zi Wang
- grid.412523.30000 0004 0386 9086Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011 China
| | - Xiao-Xue Chen
- grid.412523.30000 0004 0386 9086Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011 China
| | - Qi Zeng
- grid.415002.20000 0004 1757 8108Department of Plastic Surgery, Jiangxi Province People’s Hospital, Nanchang, China
| | - Chuan-Chang Dai
- grid.412523.30000 0004 0386 9086Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011 China
| | - Jiao Wei
- grid.412523.30000 0004 0386 9086Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011 China
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW The periosteum, the outer layer of bone, is a major source of skeletal stem/progenitor cells (SSPCs) for bone repair. Here, we discuss recent findings on the characterization, role, and regulation of periosteal SSPCs (pSSPCs) during bone regeneration. RECENT FINDINGS Several markers have been described for pSSPCs but lack tissue specificity. In vivo lineage tracing and transcriptomic analyses have improved our understanding of pSSPC functions during bone regeneration. Bone injury activates pSSPCs that migrate, proliferate, and have the unique potential to form both bone and cartilage. The injury response of pSSPCs is controlled by many signaling pathways including BMP, FGF, Notch, and Wnt, their metabolic state, and their interactions with the blood clot, nerve fibers, blood vessels, and macrophages in the fracture environment. Periosteal SSPCs are essential for bone regeneration. Despite recent advances, further studies are required to elucidate pSSPC heterogeneity and plasticity that make them a central component of the fracture healing process and a prime target for clinical applications.
Collapse
Affiliation(s)
- Simon Perrin
- Univ Paris Est Creteil, INSERM, IMRB, F-94010, Creteil, France
| | - Céline Colnot
- Univ Paris Est Creteil, INSERM, IMRB, F-94010, Creteil, France.
| |
Collapse
|
7
|
Woloszyk A, Tuong ZK, Perez L, Aguilar L, Bankole AI, Evans CH, Glatt V. Fracture hematoma micro-architecture influences transcriptional profile and plays a crucial role in determining bone healing outcomes. BIOMATERIALS ADVANCES 2022; 139:213027. [PMID: 35882120 DOI: 10.1016/j.bioadv.2022.213027] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/27/2022] [Accepted: 07/08/2022] [Indexed: 06/15/2023]
Abstract
The hematoma that forms between broken fragments of bone serves as a natural fibrin scaffold, and its removal from the defect site delays bone healing. The hypothesis of this study is that the microarchitectural and mechanical properties of the initially formed hematoma has a significant effect on the regulation of the biological process, which ultimately determines the outcome of bone healing. To mimic three healing conditions in the rat femur (normal, delayed, and non-healing bone defects), three different defect sizes of 0.5, 1.5, and 5.0 mm, are respectively used. The analysis of 3-day-old hematomas demonstrates clear differences in fibrin clot micro-architecture in terms of fiber diameter, fiber density, and porosity of the formed fibrin network, which result in different mechanical properties (stiffness) of the hematoma in each model. Those differences directly affect the biological processes involved. Specifically, RNA-sequencing reveals almost 700 differentially expressed genes between normally healing and non-healing defects, including significantly up-regulated essential osteogenic genes in normally healing defects, also differences in immune cell populations, activated osteogenic transcriptional regulators as well as potential novel marker genes. Most importantly, this study demonstrates that the healing outcome has already been determined during the hematoma phase of bone healing, three days post-surgery.
Collapse
Affiliation(s)
- Anna Woloszyk
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| | - Zewen K Tuong
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba 4102, QLD, Australia; Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK; Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK.
| | - Louis Perez
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| | - Leonardo Aguilar
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| | - Abraham I Bankole
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| | - Christopher H Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester 55902, MN, USA.
| | - Vaida Glatt
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| |
Collapse
|
8
|
Mehta AS, Deshpande P, Chimata AV, Tsonis PA, Singh A. Newt regeneration genes regulate Wingless signaling to restore patterning in Drosophila eye. iScience 2021; 24:103166. [PMID: 34746690 PMCID: PMC8551474 DOI: 10.1016/j.isci.2021.103166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/02/2021] [Accepted: 09/21/2021] [Indexed: 12/21/2022] Open
Abstract
Newts utilize their unique genes to restore missing parts by strategic regulation of conserved signaling pathways. Lack of genetic tools poses challenges to determine the function of such genes. Therefore, we used the Drosophila eye model to demonstrate the potential of 5 unique newt (Notophthalmus viridescens) gene(s), viropana1-viropana5 (vna1-vna5), which were ectopically expressed in L 2 mutant and GMR-hid, GMR-GAL4 eye. L 2 exhibits the loss of ventral half of early eye and head involution defective (hid) triggers cell-death during later eye development. Surprisingly, newt genes significantly restore missing photoreceptor cells both in L 2 and GMR>hid background by upregulating cell-proliferation and blocking cell-death, regulating evolutionarily conserved Wingless (Wg)/Wnt signaling pathway and exhibit non-cell-autonomous rescues. Further, Wg/Wnt signaling acts downstream of newt genes. Our data highlights that unique newt proteins can regulate conserved pathways to trigger a robust restoration of missing photoreceptor cells in Drosophila eye model with weak restoration capability.
Collapse
Affiliation(s)
| | | | | | | | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
- Premedical Program, University of Dayton, Dayton, USA
- Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, USA
- The Integrative Science and Engineering Center, University of Dayton, Dayton, OH 45469, USA
- Center for Genomic Advocacy (TCGA), Indiana State University, Terre Haute, IN, USA
| |
Collapse
|
9
|
Wnt/ β-Catenin Pathway Balances Scaffold Degradation and Bone Formation in Tissue-Engineered Laminae. Stem Cells Int 2021; 2021:8359582. [PMID: 34552633 PMCID: PMC8452400 DOI: 10.1155/2021/8359582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 08/23/2021] [Indexed: 01/02/2023] Open
Abstract
Tissue engineering provides a promising way for the regeneration of artificial vertebral laminae. Previous studies have confirmed the feasibility of reconstructing vertebral laminae via hydroxyapatite-collagen I scaffolds and mesenchymal stromal cells. However, there were no studies exploring the degradation of hydroxyapatite-collagen I scaffolds and the function of Wnt/β-catenin pathway in the process. In this study, tissue-engineered laminae (TEL) were constructed by nanohydroxyapatite/collagen I scaffolds and umbilical cord Wharton's Jelly mesenchymal stromal cells (WJ-MSCs). Cell attachment was observed by scanning electron microscopy, and cell viability was confirmed by Live/Dead staining. The rat models were randomly divided into control and β-catenin inhibition groups. Vertebral lamina defect rat models were made on the fifth lumbar vertebrate, and TEL was implanted into the defect site. After 14 weeks, the newborn laminae were harvested for microcomputed tomography, histology, or transcriptional profile analysis. We found that, for the control group, the newborn lamina formation matched with the scaffold degradation and complete newborn laminae formed at the 14th week; for the β-catenin inhibition group, the scaffold degradation rate overrated the lamina formation and no complete artificial laminae were formed at the 14th week. In addition, the osteoclastic genes, such as Cathepsin K or RANKL, in the control groups were significantly lower than the β-catenin inhibition group, and the antiosteoclastic gene, OPG, in the control group was significantly higher than the β-catenin inhibition group. In conclusion, inhibition of Wnt/β-catenin pathway led to speedy scaffold degradation and deferred artificial lamina formation. Wnt/β-catenin pathway played a critical role in maintaining the balance between scaffold degradation and bone formation in the process of vertebral lamina reconstruction.
Collapse
|
10
|
Mihara A, Yukata K, Seki T, Iwanaga R, Nishida N, Fujii K, Nagao Y, Sakai T. Effects of sclerostin antibody on bone healing. World J Orthop 2021; 12:651-659. [PMID: 34631449 PMCID: PMC8472444 DOI: 10.5312/wjo.v12.i9.651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/12/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
Promoting bone healing after a fracture has been a frequent subject of research. Recently, sclerostin antibody (Scl-Ab) has been introduced as a new anabolic agent for the treatment of osteoporosis. Scl-Ab activates the canonical Wnt (cWnt)-β-catenin pathway, leading to an increase in bone formation and decrease in bone resorption. Because of its rich osteogenic effects, preclinically, Scl-Ab has shown positive effects on bone healing in rodent models; researchers have reported an increase in bone mass, mechanical strength, histological bone formation, total mineralized callus volume, bone mineral density, neovascularization, proliferating cell nuclear antigen score, and bone morphogenic protein expression at the fracture site after Scl-Ab administration. In addition, in a rat critical-size femoral-defect model, the Scl-Ab-treated group demonstrated a higher bone healing rate. On the other hand, two clinical reports have researched Scl-Ab in bone healing and failed to show positive effects in the femur and tibia. This review discusses why Scl-Ab appears to be effective in animal models of fracture healing and not in clinical cases.
Collapse
Affiliation(s)
- Atsushi Mihara
- Department of Orthopedic Surgery, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| | - Kiminori Yukata
- Department of Orthopedic Surgery, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| | - Toshihiro Seki
- Department of Orthopedic Surgery, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| | - Ryuta Iwanaga
- Department of Orthopedic Surgery, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| | - Norihiro Nishida
- Department of Orthopedic Surgery, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| | - Kenzo Fujii
- Department of Orthopedic Surgery, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| | - Yuji Nagao
- Department of Orthopedic Surgery, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| | - Takashi Sakai
- Department of Orthopedic Surgery, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan
| |
Collapse
|
11
|
Fowler TW, Mitchell TL, Janda CY, Xie L, Tu S, Chen H, Zhang H, Ye J, Ouyang B, Yuan TZ, Lee SJ, Newman M, Tripuraneni N, Rego ES, Mutha D, Dilip A, Vuppalapaty M, Baribault H, Yeh WC, Li Y. Development of selective bispecific Wnt mimetics for bone loss and repair. Nat Commun 2021; 12:3247. [PMID: 34059688 PMCID: PMC8167098 DOI: 10.1038/s41467-021-23374-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
The Wnt signaling pathway is intricately connected with bone mass regulation in humans and rodent models. We designed an antibody-based platform that generates potent and selective Wnt mimetics. Using this platform, we engineer bi-specific Wnt mimetics that target Frizzled and low-density lipoprotein receptor-related proteins and evaluate their effects on bone accrual in murine models. These synthetic Wnt agonists induce rapid and robust bone building effects, and correct bone mass deficiency and bone defects in various disease models, including osteoporosis, aging, and long bone fracture. Furthermore, when these Wnt agonists are combined with antiresorptive bisphosphonates or anti-sclerostin antibody therapies, additional bone accrual/maintenance effects are observed compared to monotherapy, which could benefit individuals with severe and/or acute bone-building deficiencies. Our data support the continued development of Wnt mimetics for the treatment of diseases of low bone mineral density, including osteoporosis.
Collapse
Affiliation(s)
| | | | - Claudia Y Janda
- Surrozen, Inc., South San Francisco, CA, USA.,Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Liqin Xie
- Surrozen, Inc., South San Francisco, CA, USA
| | | | - Hui Chen
- Surrozen, Inc., South San Francisco, CA, USA
| | - Haili Zhang
- Surrozen, Inc., South San Francisco, CA, USA
| | - Jingjing Ye
- Surrozen, Inc., South San Francisco, CA, USA
| | | | - Tom Z Yuan
- Surrozen, Inc., South San Francisco, CA, USA
| | | | | | | | | | - Devin Mutha
- Surrozen, Inc., South San Francisco, CA, USA
| | | | | | | | | | - Yang Li
- Surrozen, Inc., South San Francisco, CA, USA.
| |
Collapse
|
12
|
McGowan LM, Kague E, Vorster A, Newham E, Cross S, Hammond CL. Wnt16 Elicits a Protective Effect Against Fractures and Supports Bone Repair in Zebrafish. JBMR Plus 2021; 5:e10461. [PMID: 33778326 PMCID: PMC7990157 DOI: 10.1002/jbm4.10461] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/09/2020] [Accepted: 01/03/2021] [Indexed: 12/23/2022] Open
Abstract
Bone homeostasis is a dynamic, multicellular process that is required throughout life to maintain bone integrity, prevent fracture, and respond to skeletal damage. WNT16 has been linked to bone fragility and osteoporosis in human genome wide‐association studies, as well as the functional hematopoiesis of leukocytes in vivo. However, the mechanisms by which WNT16 promotes bone health and repair are not fully understood. In this study, CRISPR‐Cas9 was used to generate mutant zebrafish lacking Wnt16 (wnt16−/−) to study its effect on bone dynamically. The wnt16 mutants displayed variable tissue mineral density (TMD) and were susceptible to spontaneous fractures and the accumulation of bone calluses at an early age. Fractures were induced in the lepidotrichia of the caudal fins of wnt16−/− and WT zebrafish; this model was used to probe the mechanisms by which Wnt16 regulates skeletal and immune cell dynamics in vivo. In WT fins, wnt16 expression increased significantly during the early stages for bone repair. Mineralization of bone during fracture repair was significantly delayed in wnt16 mutants compared with WT zebrafish. Surprisingly, there was no evidence that the recruitment of innate immune cells to fractures or soft callus formation was altered in wnt16 mutants. However, osteoblast recruitment was significantly delayed in wnt16 mutants postfracture, coinciding with precocious activation of the canonical Wnt signaling pathway. In situ hybridization suggests that canonical Wnt‐responsive cells within fractures are osteoblast progenitors, and that osteoblast differentiation during bone repair is coordinated by the dynamic expression of runx2a and wnt16. This study highlights zebrafish as an emerging model for functionally validating osteoporosis–associated genes and investigating fracture repair dynamically in vivo. Using this model, it was found that Wnt16 protects against fracture and supports bone repair, likely by modulating canonical Wnt activity via runx2a to facilitate osteoblast differentiation and bone matrix deposition. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Lucy M McGowan
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Erika Kague
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Alistair Vorster
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Elis Newham
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Stephen Cross
- Wolfson Bioimaging Facility University of Bristol Bristol UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| |
Collapse
|
13
|
Komatsu DE, Duque E, Hadjiargyrou M. MicroRNAs and fracture healing: Pre-clinical studies. Bone 2021; 143:115758. [PMID: 33212318 PMCID: PMC7769985 DOI: 10.1016/j.bone.2020.115758] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/28/2022]
Abstract
During the past several years, pre-clinical experiments have established that microRNAs (miRNAs), small non-coding RNAs, serve as key regulatory molecules of fracture healing. Their easy modulation with agonists and antagonists make them highly desirable targets for future therapeutic strategies, especially for pathophysiologic fractures that either do not heal (nonunions) or are delayed. It is now well documented that these problematic fractures lead to human suffering and impairment of life quality. Additionally, financial difficulties are also encountered as work productivity decreases and income is reduced. Moreover, targeting miRNAs may also be an avenue to enhancing normal physiological fracture healing. Herein we present the most current knowledge of the involvement of miRNAs during fracture healing in pre-clinical studies. Following a brief description on the nature of miRNAs and of the fracture healing process, we present data from studies focusing specifically, on miRNA regulation of osteoblast differentiation and osteogenesis (within the context of known signaling pathways), chondrocytes, angiogenesis, and apoptosis, all critical to successful bone repair. Further, we also discuss miRNAs and exosomes. We hope that this manuscript serves as a comprehensive review that will facilitate basic/translational scientists in the orthopaedic arena to realize and further decipher the biological and future therapeutic impact of these small regulatory RNA molecules, especially as they relate to the molecular events of each of the major phases of fracture healing.
Collapse
Affiliation(s)
- David E Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, United States of America
| | - Edie Duque
- Department of Orthopaedics and Rehabilitation, Stony Brook University, United States of America
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, United States of America.
| |
Collapse
|
14
|
Li Y, Liu G, Xiao F, Gu W, Gao Z, Wu Y, Wang P, Shi M, Yang M, Zhong Z, Liu B. Dual Role of Caveolin-1 in β-Catenin Signaling During Fracture Healing Induced by Low-Intensity Pulsed Ultrasound in Rabbits. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We did this research to observe the effect of LIPUS on long bone fracture repair and caveolin-1, β-catenin signaling expression in the radius defects of rabbits, to explore its possible molecular mechanisms. 24 male New Zealand rabbits with bilateral radial bone defects
were divided into 4 groups randomly, n = 6. The right side had daily LIPUS exposure for 20 minutes, while the left received sham treatment. After 7, 14, 21, 28 days, respectively, fracture healing was observed by X-ray imaging and Dual Energy X-ray Absorptiometry (DXA) scan, specimens
were harvested for histology, immunohistochemistry, and gene expression analysis. We found that LIPUS brought forward endochondral ossification, increased the bone callus size without changes in Bone Mineral Density (BMD). The caveolin-1 expression increased first then decreased, while the
β-catenin kept growing during the process. These demonstrated that caveolin-1 participated in fracture healing accelerated by LIPUS, which was speculated to play a dual role in β-catenin signaling expression.
Collapse
Affiliation(s)
- Yun Li
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guanghua Liu
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Feng Xiao
- Department of Pathology, Shanghai Seventh People’s Hospital, Shanghai 200137, China
| | - Wenqin Gu
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhengdong Gao
- Department of Radiology, Fenglin Community Health Service Center, Xuhui District, Shanghai 200030, China
| | - Yiming Wu
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ping Wang
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mingfang Shi
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mingzhen Yang
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zongye Zhong
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bangzhong Liu
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
Schupbach D, Comeau-Gauthier M, Harvey E, Merle G. Wnt modulation in bone healing. Bone 2020; 138:115491. [PMID: 32569871 DOI: 10.1016/j.bone.2020.115491] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022]
Abstract
Genetic studies have been instrumental in the field of orthopaedics for finding tools to improve the standard management of fractures and delayed unions. The Wnt signaling pathway that is crucial for development and maintenance of many organs also has a very promising pathway for enhancement of bone regeneration. The Wnt pathway has been shown to have a direct effect on stem cells during bone regeneration, making Wnt a potential target to stimulate bone repair after trauma. A more complete view of how Wnt influences animal bone regeneration has slowly come to light. This review article provides an overview of studies done investigating the modulation of the canonical Wnt pathway in animal bone regeneration models. This not only includes a summary of the recent work done elucidating the roles of Wnt and β-catenin in fracture healing, but also the results of thirty transgenic studies, and thirty-eight pharmacological studies. Finally, we discuss the discontinuation of sclerostin clinical trials, ongoing clinical trials with lithium, the results of Dkk antibody clinical trials, the shift into combination therapies and the future opportunities to enhance bone repair and regeneration through the modulation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Drew Schupbach
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Experimental Surgery, Faculty of Medicine, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A7-117, Montreal, Québec H3G 1A4, Canada.
| | - Marianne Comeau-Gauthier
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Experimental Surgery, Faculty of Medicine, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A7-117, Montreal, Québec H3G 1A4, Canada.
| | - Edward Harvey
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada.
| | - Geraldine Merle
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Department of Chemical Engineering, Polytechnique Montreal, 2500, chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada.
| |
Collapse
|
16
|
Mehta AS, Singh A. Insights into regeneration tool box: An animal model approach. Dev Biol 2019; 453:111-129. [PMID: 30986388 PMCID: PMC6684456 DOI: 10.1016/j.ydbio.2019.04.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 12/20/2022]
Abstract
For ages, regeneration has intrigued countless biologists, clinicians, and biomedical engineers. In recent years, significant progress made in identification and characterization of a regeneration tool kit has helped the scientific community to understand the mechanism(s) involved in regeneration across animal kingdom. These mechanistic insights revealed that evolutionarily conserved pathways like Wnt, Notch, Hedgehog, BMP, and JAK/STAT are involved in regeneration. Furthermore, advancement in high throughput screening approaches like transcriptomic analysis followed by proteomic validations have discovered many novel genes, and regeneration specific enhancers that are specific to highly regenerative species like Hydra, Planaria, Newts, and Zebrafish. Since genetic machinery is highly conserved across the animal kingdom, it is possible to engineer these genes and regeneration specific enhancers in species with limited regeneration properties like Drosophila, and mammals. Since these models are highly versatile and genetically tractable, cross-species comparative studies can generate mechanistic insights in regeneration for animals with long gestation periods e.g. Newts. In addition, it will allow extrapolation of regenerative capabilities from highly regenerative species to animals with low regeneration potential, e.g. mammals. In future, these studies, along with advancement in tissue engineering applications, can have strong implications in the field of regenerative medicine and stem cell biology.
Collapse
Affiliation(s)
- Abijeet S Mehta
- Department of Biology, University of Dayton, Dayton, OH, 45469, USA
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH, 45469, USA; Premedical Program, University of Dayton, Dayton, OH, 45469, USA; Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, 45469, USA; The Integrative Science and Engineering Center, University of Dayton, Dayton, OH, 45469, USA; Center for Genomic Advocacy (TCGA), Indiana State University, Terre Haute, IN, USA.
| |
Collapse
|
17
|
Mehta AS, Luz-Madrigal A, Li JL, Tsonis PA, Singh A. Comparative transcriptomic analysis and structure prediction of novel Newt proteins. PLoS One 2019; 14:e0220416. [PMID: 31419228 PMCID: PMC6697330 DOI: 10.1371/journal.pone.0220416] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/15/2019] [Indexed: 01/25/2023] Open
Abstract
Notophthalmus viridescens (Red-spotted Newt) possess amazing capabilities to regenerate their organs and other tissues. Previously, using a de novo assembly of the newt transcriptome combined with proteomic validation, our group identified a novel family of five protein members expressed in adult tissues during regeneration in Notophthalmus viridescens. The presence of a putative signal peptide suggests that all these proteins are secretory in nature. Here we employed iterative threading assembly refinement (I-TASSER) server to generate three-dimensional structure of these novel Newt proteins and predicted their function. Our data suggests that these proteins could act as ion transporters, and be involved in redox reaction(s). Due to absence of transgenic approaches in N. viridescens, and conservation of genetic machinery across species, we generated transgenic Drosophila melanogaster to misexpress these genes. Expression of 2775 transcripts were compared between these five newly identified Newt genes. We found that genes involved in the developmental process, cell cycle, apoptosis, and immune response are among those that are highly enriched. To validate the RNA Seq. data, expression of six highly regulated genes were verified using real time Quantitative Polymerase Chain Reaction (RT-qPCR). These graded gene expression patterns provide insight into the function of novel protein family identified in Newt, and layout a map for future studies in the field.
Collapse
Affiliation(s)
- Abijeet Singh Mehta
- Department of Biology, University of Dayton, Dayton, Ohio, United States of America
| | - Agustin Luz-Madrigal
- Department of Biology, University of Dayton, Dayton, Ohio, United States of America
| | - Jian-Liang Li
- Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida, United States of America
| | - Panagiotis A Tsonis
- Department of Biology, University of Dayton, Dayton, Ohio, United States of America
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, Ohio, United States of America
- Premedical Program, University of Dayton, Dayton, Ohio, United States of America
- Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, Ohio, United States of America
- The Integrative Science and Engineering Center, University of Dayton, Dayton, Ohio, United States of America
- Center for Genomic Advocacy (TCGA), Indiana State University, Terre Haute, Indiana, United States of America
| |
Collapse
|
18
|
Abstract
Poorly controlled diabetes with comorbid manifestations negatively affects outcomes in lower extremity trauma, increasing the risk of short-term and long-term complications. Management strategies of patients with diabetes that experience lower extremity trauma should also include perioperative management of hyperglycemia to reduce adverse and serious adverse events.
Collapse
Affiliation(s)
- George T Liu
- Orthopaedic Surgery, University of Texas Southwestern Medical Center, 1801 Inwood Road, Dallas, TX 75390-8883, USA; Foot and Ankle Service, Orthopaedic Surgery, Parkland Memorial Hospital, Level 1 Trauma Center, 5200 Harry Hines Boulevard, Dallas, TX 75235, USA.
| | - Drew T Sanders
- Orthopaedic Surgery, University of Texas Southwestern Medical Center, 1801 Inwood Road, Dallas, TX 75390-8883, USA; Orthopaedic Trauma Service, Parkland Memorial Hospital, Level 1 Trauma Center, 5200 Harry Hines Boulevard, Dallas, TX 75235, USA
| | - Katherine M Raspovic
- Orthopaedic Surgery, University of Texas Southwestern Medical Center, 1801 Inwood Road, Dallas, TX 75390-8883, USA; Foot and Ankle Service, Orthopaedic Surgery, Parkland Memorial Hospital, Level 1 Trauma Center, 5200 Harry Hines Boulevard, Dallas, TX 75235, USA
| | - Dane K Wukich
- Orthopaedic Surgery, University of Texas Southwestern Medical Center, 1801 Inwood Road, Dallas, TX 75390-8883, USA; Foot and Ankle Service, Orthopaedic Surgery, Parkland Memorial Hospital, Level 1 Trauma Center, 5200 Harry Hines Boulevard, Dallas, TX 75235, USA
| |
Collapse
|
19
|
Sarmah S, Curtis C, Mahin J, Farrell M, Engler TA, Sanchez-Felix MV, Sato M, Ma YL, Chu S, Marrs JA. The Glycogen Synthase Kinase-3β Inhibitor LSN 2105786 Promotes Zebrafish Fin Regeneration. Biomedicines 2019; 7:biomedicines7020030. [PMID: 31010223 PMCID: PMC6630808 DOI: 10.3390/biomedicines7020030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/12/2019] [Accepted: 04/14/2019] [Indexed: 11/16/2022] Open
Abstract
The Wnt pathway has been shown to regulate bone homeostasis and to influence some bone disease states. We utilized a zebrafish model system to study the effects of a synthetic, orally bioavailable glycogen synthase kinase-3β (GSK3β) inhibitor LSN 2105786, which activates Wnt signaling during bone healing and embryogenesis. GSK3β inhibitor treatment was used to phenocopy GSK3β morpholino oligonucleotide (MO) knockdown in zebrafish embryos. Human and zebrafish synthetic mRNA injection were similarly effective at rescue of GSK3β MO knockdown. During caudal fin regeneration, bony rays are the first structure to differentiate in zebrafish fins, providing a useful model to study bone healing. Caudal fin regeneration experiments were conducted using various concentrations of a GSK3β inhibitor, examining duration and concentration dependence on regenerative outgrowth. Experiments revealed continuous low concentration (4-5 nM) treatment to be more effective at increasing regeneration than intermittent dosing. Higher concentrations inhibited fin growth, perhaps by excessive stimulation of differentiation programs. Increased Wnt responsive gene expression and differentiation were observed in response to GSK3b inhibitor treatment. Activating Wnt signaling also increased cell proliferation and osteoblast differentiation in fin regenerates. Together, these data indicate that bone healing in zebrafish fin regeneration was improved by activating Wnt signaling using GSK3b inhibitor treatment. In addition, caudal fin regeneration is useful to evaluate dose-dependent pharmacological efficacy in bone healing, various dosing regimens and possible toxicological effects of compounds.
Collapse
Affiliation(s)
- Swapnalee Sarmah
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | - Courtney Curtis
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | - Jennifer Mahin
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | - Mark Farrell
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | | | - Manuel V Sanchez-Felix
- Lilly Research Laboratories, Indianapolis, IN 46225, USA.
- Novartis Institute for BioMedical Research, Cambridge, MA 02139, USA.
| | - Masahiko Sato
- Lilly Research Laboratories, Indianapolis, IN 46225, USA.
| | | | - Shaoyou Chu
- Lilly Research Laboratories, Indianapolis, IN 46225, USA.
- Molecular Templates, Austin, TX 78729, USA.
| | - James A Marrs
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| |
Collapse
|
20
|
Bahney CS, Zondervan RL, Allison P, Theologis A, Ashley JW, Ahn J, Miclau T, Marcucio RS, Hankenson KD. Cellular biology of fracture healing. J Orthop Res 2019; 37:35-50. [PMID: 30370699 PMCID: PMC6542569 DOI: 10.1002/jor.24170] [Citation(s) in RCA: 351] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/27/2018] [Indexed: 02/04/2023]
Abstract
The biology of bone healing is a rapidly developing science. Advances in transgenic and gene-targeted mice have enabled tissue and cell-specific investigations of skeletal regeneration. As an example, only recently has it been recognized that chondrocytes convert to osteoblasts during healing bone, and only several years prior, seminal publications reported definitively that the primary tissues contributing bone forming cells during regeneration were the periosteum and endosteum. While genetically modified animals offer incredible insights into the temporal and spatial importance of various gene products, the complexity and rapidity of healing-coupled with the heterogeneity of animal models-renders studies of regenerative biology challenging. Herein, cells that play a key role in bone healing will be reviewed and extracellular mediators regulating their behavior discussed. We will focus on recent studies that explore novel roles of inflammation in bone healing, and the origins and fates of various cells in the fracture environment. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Chelsea S. Bahney
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Robert L. Zondervan
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | - Patrick Allison
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
| | - Alekos Theologis
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Jason W. Ashley
- Department of Biology, Eastern Washington University, Cheney, Washington
| | - Jaimo Ahn
- Department of Biology, Eastern Washington University, Cheney, Washington
| | - Theodore Miclau
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Ralph S. Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
21
|
Alzahrani MM, Makhdom AM, Rauch F, Lauzier D, Kotsiopriftis M, Ghadakzadeh S, Hamdy RC. Assessment of the effect of systemic delivery of sclerostin antibodies on Wnt signaling in distraction osteogenesis. J Bone Miner Metab 2018. [PMID: 28647818 DOI: 10.1007/s00774-017-0847-2] [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] [Indexed: 12/13/2022]
Abstract
Sclerostin is a known inhibitor of the Wnt signaling pathway which is involved in osteogenesis and, when inactivated, stimulates bone formation. To our knowledge, this effect has not been studied in the context of distraction osteogenesis (DO). Tibial DO was conducted on a total of 24 wild-type mice, which were then divided into 2 groups-a saline injection group (control) and an anti-sclerostin (Scl-Ab) injection group (treatment). The mice in the treatment group received 100 mg/kg intravenous injections of the antibody weekly until killing. The 12 mice in each group were subdivided into four time points according to post-osteotomy time of killing-11 days (mid-distraction), 17 days (late distraction), 34 days (mid-consolidation) and 51 days (late consolidation), with 3 mice per subgroup. After killing, the tibia specimens were collected for immunohistochemical analysis. Our results show that the group injected with anti-sclerostin had an earlier peak (day 11) in the distraction phase of the osteogenic molecules involved in the Wnt signaling pathway in comparison to the placebo group. In addition, downregulation of the inhibitors of this pathway was noted in the treatment group when compared with the placebo group. Furthermore, LRP-5 showed a significant increase in expression in the treatment group. Sclerostin inhibition has a significant effect on the DO process through its effect on the Wnt pathway. This effect was evident through the decreased effect of sclerostin on LRP-5 and earlier upregulation of the osteogenic molecules involved in this pathway.
Collapse
Affiliation(s)
- Mohammad M Alzahrani
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, 1003 Decarie Blvd, Montreal, QC, H4A 0A9, Canada.
- Department of Orthopaedic Surgery, University of Dammam, Dammam, Saudi Arabia.
| | - Asim M Makhdom
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, 1003 Decarie Blvd, Montreal, QC, H4A 0A9, Canada
- Department of Orthopaedic Surgery, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Frank Rauch
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, 1003 Decarie Blvd, Montreal, QC, H4A 0A9, Canada
| | - Dominique Lauzier
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, 1003 Decarie Blvd, Montreal, QC, H4A 0A9, Canada
| | - Maria Kotsiopriftis
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, 1003 Decarie Blvd, Montreal, QC, H4A 0A9, Canada
| | - Saber Ghadakzadeh
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, 1003 Decarie Blvd, Montreal, QC, H4A 0A9, Canada
| | - Reggie C Hamdy
- Division of Orthopaedic Surgery, Shriners Hospital for Children, Montreal Children Hospital, McGill University, 1003 Decarie Blvd, Montreal, QC, H4A 0A9, Canada
| |
Collapse
|
22
|
Li Z, Xu Z, Duan C, Liu W, Sun J, Han B. Role of TCF/LEF Transcription Factors in Bone Development and Osteogenesis. Int J Med Sci 2018; 15:1415-1422. [PMID: 30275770 PMCID: PMC6158667 DOI: 10.7150/ijms.26741] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/29/2018] [Indexed: 11/05/2022] Open
Abstract
Bone formation occurs by two distinct mechanisms, namely, periosteal ossification and endochondral ossification. In both mechanisms, osteoblasts play an important role in the secretion and mineralization of bone-specific extracellular matrix. Differentiation and maturation of osteoblasts is a prerequisite to bone formation and is regulated by many factors. Recent experiments have shown that transcription factors play an important role in regulating osteoblast differentiation, proliferation, and function. Osteogenesis related transcription factors are the central targets and key mediators of the function of growth factors, such as cytokines. Transcription factors play a key role in the transformation of mesenchymal progenitor cells into functional osteoblasts. These transcription factors are closely linked with each other and in conjunction with bone-related signaling pathways form a complex network that regulates osteoblast differentiation and bone formation. In this paper, we discuss the structure of T-cell factor/lymphoid enhancer factor (TCF/LEF) and its role in embryonic skeletal development and the crosstalk with related signaling pathways and factors.
Collapse
Affiliation(s)
- Zhengqiang Li
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Jilin University, Changchun 130021, China.,Stomatological Hospital of Southern Medical University & Guangdong Provincial Stomatological Hospital, Guangzhou 510280, China
| | - Zhimin Xu
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| | - Congcong Duan
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| | - Weiwei Liu
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| | - Jingchun Sun
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| | - Bing Han
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| |
Collapse
|
23
|
Wang X, Luo E, Bi R, Ye B, Hu J, Zou S. Wnt/β-catenin signaling is required for distraction osteogenesis in rats. Connect Tissue Res 2018; 59:45-54. [PMID: 28346008 DOI: 10.1080/03008207.2017.1300154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OVERVIEW The Wnt signaling pathway plays crucial roles in embryonic skeletal development and postnatal bone regeneration. However, mechanisms of Wnt signaling functioning in distraction osteogenesis (DO) haven't been well characterized. MATERIALS AND METHODS We established a DO model using Sprague-Dawley rat tibia. And a Wnt signaling blocking agent, recombinant rat Dickkopf-related protein 1 (rrDkk1), was locally applied in the distracted gap to study the role of Wnt signaling during DO process. Animals in the experimental group received rrDkk1 injections (dose = 25 μg/kg) once daily during distraction period and every third day during consolidation stage (n = 48). Animals in the control group received saline under the same injection strategy (n = 48). Animals at different time points during DO process (1, 3, 6, 12 days after distraction, 10 days and 6 weeks after consolidation) were killed and tissues in the distraction region were harvested for radiography, dual energy X-ray absorptiometry, micro-computed tomography (micro-CT), and histological analyses. RESULTS Most Wnt ligands, cofactors, receptors, and antagonists were widely expressed in the distraction callus and were significantly upregulated during DO process. After rrDkk1 administration, the majority of these factors were downregulated at the mRNA level, except sFRP and GSK-3β. At the protein level, both β-catenin and Lef-1 were also suppressed by rrDkk1. In the long term, restricted bone healing was observed in the distracted callus in the rrDkk1 injection group. These findings were confirmed by histological and micro-CT analyses. CONCLUSIONS Our findings suggest that Wnt signaling participates in the process of DO, and clinical therapeutic approaches of DO may do well to avoid Wnt pathway suppression.
Collapse
Affiliation(s)
- Xuemei Wang
- a State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases , West China Hospital of Stomatology, Sichuan University , Chengdu , China
| | - En Luo
- a State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases , West China Hospital of Stomatology, Sichuan University , Chengdu , China
| | - Ruiye Bi
- a State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases , West China Hospital of Stomatology, Sichuan University , Chengdu , China
| | - Bin Ye
- a State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases , West China Hospital of Stomatology, Sichuan University , Chengdu , China
| | - Jing Hu
- a State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases , West China Hospital of Stomatology, Sichuan University , Chengdu , China
| | - Shujuan Zou
- a State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases , West China Hospital of Stomatology, Sichuan University , Chengdu , China
| |
Collapse
|
24
|
Extracorporeal Shock Wave Rebuilt Subchondral Bone In Vivo and Activated Wnt5a/Ca 2+ Signaling In Vitro. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1404650. [PMID: 29164146 PMCID: PMC5661093 DOI: 10.1155/2017/1404650] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 08/14/2017] [Accepted: 08/29/2017] [Indexed: 12/27/2022]
Abstract
Background This study aimed to identify the optimal extracorporeal shock wave (ESW) intensity and to investigate its effect on subchondral bone rebuilt in vivo and Wnt5a/Ca2+ signaling in vitro using an osteoarthritis (OA) rat model and bone marrow mesenchymal stem cells (BMMSCs), respectively. Methods OA rats treated with (OA + ESW group) or without (OA group) ESW (n = 12/group) were compared with healthy controls (control group, n = 12). Gait patterns and subchondral trabecular bone changes were measured. Western blot and quantitative real-time polymerase chain reaction detected protein expression and gene transcription, respectively. Results The gait disturbances of OA + ESW group were significantly improved compared with the OA group at 6th and 8th weeks. The micro-CT analysis indicated that the BMD, BSV/BV, BV/TV, Tr.S, and Tr.Th are significantly different between OA group and OA + ESW group. Expression of Wnt5a was increased rapidly after ESW treatment at 0.6 bar and peaked after 30 min. Conclusions ESW were positive for bone remodeling in joint tibial condyle subchondral bone of OA rat. ESW prevented histological changes in OA and prevented gait disturbance associated with OA progression. Optimal intensity of ESW induced changes in BMMSCs via activation of the Wnt5a/Ca2+ signaling pathway.
Collapse
|
25
|
Ling Z, Wu L, Shi G, Chen L, Dong Q. Increased Runx2 expression associated with enhanced Wnt signaling in PDLLA internal fixation for fracture treatment. Exp Ther Med 2017; 13:2085-2093. [PMID: 28565812 DOI: 10.3892/etm.2017.4216] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 11/10/2016] [Indexed: 01/19/2023] Open
Abstract
Poly-D-L lactide (PDLLA) biodegradable implants to heal fractures are widely applied in orthopedic surgeries. However, whether the process of fracture healing is regulated differently when PDLLA is used compared with traditional metal materials remains unclear. Runt-related transcription factor 2 (Runx2) and canonical Wnt signaling are essential and may interact reciprocally in the regulation of osteogenesis during bone repair. In the present study, a rat femoral open osteotomy model was used to compare the curative efficacy of a PDLLA rod and Kirschner wire under intramedullary fixation for fracture treatment. The dynamic expression of Runx2 and key components of the canonical Wnt signaling in callus tissue during fracture healing was also investigated. The results of the current study indicate that at weeks 4 and 6 following fixation, the callus bone structural parameters of microCT were significantly improved by PDLLA rod compared to that of Kirschner wire. In addition, at weeks 4 and 6 after fixation, the protein and mRNA expression of Runx2 and the positive regulators of canonical Wnt signaling, such as Wnts and β-catenin, were significantly increased. However, the protein and mRNA expression levels of the negative regulators of canonical Wnt signaling, such as glycogen synthase kinase-3β, were significantly decreased in callus tissue when treated with PDLLA rod compared with Kirschner wire. Collectively, these data indicate that compared to the traditional metal material, using PDLLA internal fixation for fracture treatment may further improve bone formation, which is associated with the increased expression of Runx2 and the enhancement of canonical Wnt signaling.
Collapse
Affiliation(s)
- Zhuoyan Ling
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Lei Wu
- Centers for Disease Control and Prevention of Suzhou Industrial Park, Suzhou, Jiangsu 215021, P.R. China.,School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Gaolong Shi
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Li Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qirong Dong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
26
|
Low SA, Galliford CV, Jones-Hall YL, Roy J, Yang J, Low PS, Kopeček J. Healing efficacy of fracture-targeted GSK3β inhibitor-loaded micelles for improved fracture repair. Nanomedicine (Lond) 2017; 12:185-193. [PMID: 28093944 DOI: 10.2217/nnm-2016-0340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: To evaluate the fracture healing capabilities of a GSK3β inhibitor, 6-bromoindirubin-3′-oxime, coupled with an aspartic acid octapeptide in a micellar delivery system. Materials & methods: The efficacy of the intravenously administered micelles to accelerate healing of femoral fracture in mice was evaluated. Micro-computed tomography analysis was employed to obtain bone density, total volume, relative volume, trabecular thickness and trabecular spacing.Results: Both fracture bone mineral density and volume were significantly higher in the micelle treatment groups when compared with controls. The fracture-targeted micelle demonstrates fracture-specific bone anabolism and biocompatibility in off-target tissues. Conclusion: Accelerated fracture healing in mice was achieved by targeting the GSK3β inhibitor, 6-bromoindirubin-3′-oxime, to the fracture site.
Collapse
Affiliation(s)
- Stewart A Low
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Chris V Galliford
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Yava L Jones-Hall
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Jyoti Roy
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Jiyuan Yang
- Department of Pharmaceutics & Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Jindřich Kopeček
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Pharmaceutics & Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
27
|
Significant Associations of SOX9 Gene Polymorphism and Gene Expression with the Risk of Osteonecrosis of the Femoral Head in a Han Population in Northern China. BIOMED RESEARCH INTERNATIONAL 2017; 2016:5695317. [PMID: 28090537 PMCID: PMC5174161 DOI: 10.1155/2016/5695317] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 11/08/2016] [Indexed: 01/15/2023]
Abstract
Sex determining region Y-box 9 (SOX9) is a key transcription factor involved in cartilage formation during the embryonic development stage and cartilage growth and repair after birth. To explore the roles of polymorphism and expression of the SOX9 gene in the development of osteonecrosis of the femoral head (ONFH), we analyzed the polymorphism of rs12601701 [A/G] and rs1042667 [A/C] and the serum protein expression of the SOX9 gene in 182 patients with ONFH and 179 healthy control subjects. Results revealed that the A-A haplotype of SOX9 gene as well as the GG and AA genotypes of rs12601701 was significantly associated with increased ONFH risk (P = 0.038) and the risk of bilateral hip lesions of ONFH (P = 0.009), respectively. The C-A, A-A, and A-G haplotypes were also statistically associated with the decreased and increased risk of bilateral hip lesions of ONFH (P = 0.03, P = 0.048, and P = 0.013), respectively, while the A-A haplotype closely related to the clinical stages of ONFH (P = 0.041). More importantly, the serum SOX9 protein expression of the ONFH group was greatly decreased compared to control group (P = 0.0001). Our results first showed that the gene polymorphism and gene expression of SOX9 were significantly associated with the risk and clinical phenotypes of ONFH and also indicate that the SOX9 gene may play a key role in the development of ONFH.
Collapse
|
28
|
Yi H, Ur Rehman F, Zhao C, Liu B, He N. Recent advances in nano scaffolds for bone repair. Bone Res 2016; 4:16050. [PMID: 28018707 PMCID: PMC5153570 DOI: 10.1038/boneres.2016.50] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/19/2016] [Accepted: 09/30/2016] [Indexed: 12/17/2022] Open
Abstract
Biomedical applications of nanomaterials are exponentially increasing every year due to analogy to various cell receptors, ligands, structural proteins, and genetic materials (that is, DNA). In bone tissue, nanoscale materials can provide scaffold for excellent tissue repair via mechanical stimulation, releasing of various loaded drugs and mediators, 3D scaffold for cell growth and differentiation of bone marrow stem cells to osteocytes. This review will therefore highlight recent advancements on tissue and nanoscale materials interaction.
Collapse
Affiliation(s)
- Huan Yi
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University , Nanjing, China
| | - Fawad Ur Rehman
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University , Nanjing, China
| | - Chunqiu Zhao
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University , Nanjing, China
| | - Bin Liu
- Department of Biomedical Engineering, School of Basic Medical Sciences, Nanjing Medical University , Nanjing, China
| | - Nongyue He
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing, China; Hunan Key Laboratory of Green Chemistry and Application of Biological Nanotechnology, Hunan University of Technology, Zhuzhou, China
| |
Collapse
|
29
|
Camarata T, Vasilyev A, Hadjiargyrou M. Cloning of zebrafish Mustn1 orthologs and their expression during early development. Gene 2016; 593:235-241. [DOI: 10.1016/j.gene.2016.08.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/15/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
|
30
|
Burgers TA, Vivanco JF, Zahatnansky J, Moren AJV, Mason JJ, Williams BO. Mice with a heterozygous Lrp6 deletion have impaired fracture healing. Bone Res 2016; 4:16025. [PMID: 27635281 PMCID: PMC5011612 DOI: 10.1038/boneres.2016.25] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/17/2016] [Accepted: 06/22/2016] [Indexed: 01/07/2023] Open
Abstract
Bone fracture non-unions, the failure of a fracture to heal, occur in 10%–20% of fractures and are a costly and debilitating clinical problem. The Wnt/β-catenin pathway is critical in bone development and fracture healing. Polymorphisms of linking low-density lipoprotein receptor-related protein 6 (LRP6), a Wnt-binding receptor, have been associated with decreased bone mineral density and fragility fractures, although this remains controversial. Mice with a homozygous deletion of Lrp6 have severe skeletal abnormalities and are not viable, whereas mice with a heterozygous deletion have a combinatory effect with Lrp5 to decrease bone mineral density. As fracture healing closely models embryonic skeletal development, we investigated the process of fracture healing in mice heterozygous for Lrp6 (Lrp6+/−) and hypothesized that the heterozygous deletion of Lrp6 would impair fracture healing. Mid-diaphyseal femur fractures were induced in Lrp6+/− mice and wild-type controls (Lrp6+/+). Fractures were analyzed using micro-computed tomography (μCT) scans, biomechanical testing, and histological analysis. Lrp6+/− mice had significantly decreased stiffness and strength at 28 days post fracture (PF) and significantly decreased BV/TV, total density, immature bone density, and mature area within the callus on day-14 and -21 PF; they had significantly increased empty callus area at days 14 and 21 PF. Our results demonstrate that the heterozygous deletion of Lrp6 impairs fracture healing, which suggests that Lrp6 has a role in fracture healing.
Collapse
Affiliation(s)
- Travis A Burgers
- Center for Cancer and Cell Biology, Program for Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute , Grand Rapids, MI, USA
| | - Juan F Vivanco
- Facultad de Ingenieria y Ciencias, Adolfo Ibáñez University , Viña del Mar, Chile
| | - Juraj Zahatnansky
- Center for Cancer and Cell Biology, Program for Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute , Grand Rapids, MI, USA
| | - Andrew J Vander Moren
- Padnos College of Engineering and Computing, Grand Valley State University , Grand Rapids, MI, USA
| | - James J Mason
- Center for Cancer and Cell Biology, Program for Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute , Grand Rapids, MI, USA
| | - Bart O Williams
- Center for Cancer and Cell Biology, Program for Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute , Grand Rapids, MI, USA
| |
Collapse
|
31
|
Gao J, Liu J, Yang Y, Liang J, Xie J, Li S, Zheng G, Xie L, Zhang R. Identification and expression characterization of three Wnt signaling genes in pearl oyster ( Pinctada fucata ). Comp Biochem Physiol B Biochem Mol Biol 2016; 196-197:92-101. [DOI: 10.1016/j.cbpb.2016.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 11/28/2022]
|
32
|
Hadjiargyrou M, Zhi J, Komatsu DE. Identification of the microRNA transcriptome during the early phases of mammalian fracture repair. Bone 2016; 87:78-88. [PMID: 27058875 DOI: 10.1016/j.bone.2016.03.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/10/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
Fracture repair is a complex process that involves multiple biological processes requiring spatiotemporal expression of thousands of genes. The molecular regulation of this process is not completely understood. MicroRNAs (miRNAs) regulate gene expression by promoting mRNA degradation or blocking translation. To identify miRNAs expressed during fracture repair, we generated murine bone fractures and isolated miRNA-enriched RNA from intact and post-fracture day (PFD) 1, 3, 5, 7, 11, and 14 femurs. RNA samples were individually hybridized to mouse miRNA microarrays. Results indicated that 959 (51%) miRNAs were absent while 922 (49%) displayed expression in at least one sample. Of the 922 miRNAs, 306 (33.2%) and 374 (40.6%) were up- and down-regulated, respectively, in the calluses in comparison to intact bone. Additionally, 20 (2.2%) miRNAs displayed combined up- and down-regulated expression within the time course and the remaining 222 (24%) miRNAs did not exhibit any changes between calluses and intact bone. Quantitative-PCR validated the expression of several miRNAs. Further, we identified 2048 and 4782 target genes that were unique to the up- and down-regulated miRNAs, respectively. Gene ontology and pathway enrichment analyses indicated relevant biological processes. These data provide the first complete analysis of the miRNA transcriptome during the early phases of fracture repair.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Life Sciences, Theobald Science Center, Room 420, New York Institute of Technology, Old Westbury, NY 11568-8000, USA.
| | - Jizu Zhi
- Bioinformatics Core Facility, Stony Brook University, Stony Brook, NY 11794, USA.
| | - David E Komatsu
- Department of Orthopaedics, HSC T18 Room 85, Stony Brook University, Stony Brook, NY 11794-8181, USA.
| |
Collapse
|
33
|
Bolander J, Chai YC, Geris L, Schrooten J, Lambrechts D, Roberts SJ, Luyten FP. Early BMP, Wnt and Ca(2+)/PKC pathway activation predicts the bone forming capacity of periosteal cells in combination with calcium phosphates. Biomaterials 2016; 86:106-18. [PMID: 26901484 DOI: 10.1016/j.biomaterials.2016.01.059] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 02/08/2023]
Abstract
The development of osteoinductive calcium phosphate- (CaP) based biomaterials has, and continues to be, a major focus in the field of bone tissue engineering. However, limited insight into the spatiotemporal activation of signalling pathways has hampered the optimisation of in vivo bone formation and subsequent clinical translation. To gain further knowledge regarding the early molecular events governing bone tissue formation, we combined human periosteum derived progenitor cells with three types of clinically used CaP-scaffolds, to obtain constructs with a distinct range of bone forming capacity in vivo. Protein phosphorylation together with gene expression for key ligands and target genes were investigated 24 hours after cell seeding in vitro, and 3 and 12 days post ectopic implantation in nude mice. A computational modelling approach was used to deduce critical factors for bone formation 8 weeks post implantation. The combined Ca(2+)-mediated activation of BMP-, Wnt- and PKC signalling pathways 3 days post implantation were able to discriminate the bone forming from the non-bone forming constructs. Subsequently, a mathematical model able to predict in vivo bone formation with 96% accuracy was developed. This study illustrates the importance of defining and understanding CaP-activated signalling pathways that are required and sufficient for in vivo bone formation. Furthermore, we demonstrate the reliability of mathematical modelling as a tool to analyse and deduce key factors within an empirical data set and highlight its relevance to the translation of regenerative medicine strategies.
Collapse
Affiliation(s)
- Johanna Bolander
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium
| | - Yoke Chin Chai
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Biomechanics Research Unit, University of Liege, Chemin des Chevreuils 1, BAT 52/3, 4000 Liege 1, Belgium; Biomechanics Section, KU Leuven, Celestijnenlaan 300C, Bus 2419, 3001 Leuven, Belgium
| | - Jan Schrooten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Department of Materials Engineering, KU Leuven, Kasteelpark Arenberg 44, Bus 2450, 3001 Heverlee, Belgium
| | - Dennis Lambrechts
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium
| | - Scott J Roberts
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery & Interventional Science, University College London, The Royal National Orthopaedic Hospital, Stanmore, Middlesex, HA7 4LP, United Kingdom
| | - Frank P Luyten
- Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1, Herestraat 49, Bus 813, 3000 Leuven, Belgium.
| |
Collapse
|
34
|
Abstract
Wnt-1 inducible signaling pathway-1 (WISP-1), also known as CCN-4, belongs to the connective tissue growth factor (CTGF) family. WISP-1 is primarily expressed in embryonic stem cells and is involved in adult organ development. WISP-1 participates in many cellular processes, including proliferation, differentiation, apoptosis and adhesion. In addition, WISP-1 plays an important role in diverse pathophysiological processes, such as embryonic development, inflammation, injury repairs and cancers. Recent studies showed that WISP-1 was highly correlated with tumor progression and malignant transformation, whereas it played an oncogenic role in colorectal cancer, cholangiocarcinoma, hepatocellular carcinoma and breast cancer. However, interestingly, WISP-1 exerts a tumor-suppressing role in lung and prostate cancers. WISP-1 promotes cell proliferation, adhesion, motility, invasion, metastasis and epithelial-to-mesenchymal transition via particular signaling pathways. In this review, we discussed the structure, expression profile, functions, clinical significance and potential mechanisms of WISP-1 in cancer and non-neoplastic diseases.
Collapse
Affiliation(s)
- Mengmeng Feng
- Laboratory of Surgery, the Affiliated Hospital, Inner Mongolia Medical University, Hohhot 010050, China
| | - Shuqin Jia
- Laboratory of Surgery, the Affiliated Hospital, Inner Mongolia Medical University, Hohhot 010050, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Molecular Oncology Laboratory, Peking University Cancer Hospital & Institute, Beijing 100142, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center for Molecular Diagnosis, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
35
|
Schira J, Schulte M, Döbele C, Wallner C, Abraham S, Daigeler A, Kneser U, Lehnhardt M, Behr B. Human scaphoid non-unions exhibit increased osteoclast activity compared to adjacent cancellous bone. J Cell Mol Med 2015; 19:2842-50. [PMID: 26416438 PMCID: PMC4687713 DOI: 10.1111/jcmm.12677] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 08/03/2015] [Indexed: 01/08/2023] Open
Abstract
Scaphoid bones have a high prevalence for non-union. Even with adequate treatment, bone regeneration may not occur in certain instances. Although this condition is well described, the molecular pathology of scaphoid non-unions is still poorly defined. In this study, gene expression of osteogenic and angiogenic growth and transcription factors as well as inflammatory mediators were analysed in human scaphoid non-unions and intraindividually compared to adjacent autologous cancellous bone from the distal radius. In addition, histology and immunohistochemical stainings were performed to verify qRT-PCR data. Gene expression analysis revealed a significant up-regulation of RANKL, ALP, CYCLIN D1, MMP-13, OPG, NFATc1, TGF-β and WNT5A in scaphoid non-unions. Interestingly, RANKL and NFATc1, both markers for osteoclastogenesis, were significantly induced in non-unions. Moreover, WNT5A was highly up-regulated in all non-union samples. TRAP staining confirmed the observation of induced osteoclastogenesis in non-unions. With respect to genes related to osteogenesis, alkaline phosphatase was significantly up-regulated in scaphoid non-unions. No differences were detectable for other osteogenic genes such as RUNX-2 or BMP-2. Importantly, we did not detect differences in angiogenesis between scaphoid non-unions and controls in both gene expression and immunohistochemistry. Summarized, our data indicate increased osteoclast activity in scaphoid non-unions possibly as a result of the alterations in RANKL, TGF-β and WNT5A expression levels. These data increase our understanding for the reduced bone regeneration capacity present in scaphoid non-unions and may translate into the identification of new therapeutic targets to avoid secondary damages and prevent occurrence of non-unions to scaphoid bones.
Collapse
Affiliation(s)
- Jessica Schira
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Matthias Schulte
- Department of Plastic Surgery, BG Trauma Hospital Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Carmen Döbele
- Department of Plastic Surgery, BG Trauma Hospital Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Christoph Wallner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Stephanie Abraham
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Adrien Daigeler
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Ulrich Kneser
- Department of Plastic Surgery, BG Trauma Hospital Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Marcus Lehnhardt
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Björn Behr
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
36
|
Xu H, Duan J, Ning D, Li J, Liu R, Yang R, Jiang JX, Shang P. Role of Wnt signaling in fracture healing. BMB Rep 2015; 47:666-72. [PMID: 25301020 PMCID: PMC4345510 DOI: 10.5483/bmbrep.2014.47.12.193] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Indexed: 01/08/2023] Open
Abstract
The Wnt signaling pathway is well known to play major roles in skeletal development and homeostasis. In certain aspects, fracture repair mimics the process of bone embryonic development. Thus, the importance of Wnt signaling in fracture healing has become more apparent in recent years. Here, we summarize recent research progress in the area, which may be conducive to the development of Wnt-based therapeutic strategies for bone repair. [BMB Reports 2014; 47(12): 666-672]
Collapse
Affiliation(s)
- Huiyun Xu
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, People's Republic of China
| | - Jing Duan
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, People's Republic of China
| | - Dandan Ning
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, People's Republic of China
| | - Jingbao Li
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, People's Republic of China
| | - Ruofei Liu
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, People's Republic of China
| | - Ruixin Yang
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, People's Republic of China
| | - Jean X Jiang
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, Texas 78229, the United States
| | - Peng Shang
- Key Laboratory for Space Biosciences & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, People's Republic of China
| |
Collapse
|
37
|
Li S, Yang X, Tang S, Zhang X, Feng Z, Cui S. Repair of massively defected hemi-joints using demineralized osteoarticular allografts with protected cartilage. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:227. [PMID: 26319778 DOI: 10.1007/s10856-015-5557-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 08/06/2015] [Indexed: 06/04/2023]
Abstract
Surgical replacement of massively defected joints necessarily relies on osteochondral grafts effective to both of bone and cartilage. Demineralized bone matrix (DBM) retains the osteoconductivity but destroys viable chondrocytes in the cartilage portion essential for successful restoration of defected joints. This study prepared osteochondral grafts of DBM with protected cartilage. Protected cartilage portions was characterized by cellular and molecular biology and the grafts were allogenically used for grafting. Protected cartilage showed similar histomorphological structure and protected proteins estimated by total proteins and cartilage specific proteins as in those of fresh controls when DBMs were generated in bone portions. Such grafts were successfully used for simultaneously repair of bone and cartilage in massively defected osteoarticular joints within 16 weeks post-surgery. These results present an allograft with clinical potential for simultaneous restoration of bone and cartilage in defected joints.
Collapse
Affiliation(s)
- Siming Li
- Guangzhou Institute of Traumatic Surgery, The Fourth Affiliated Hospital, Jinan University School of Medicine, 396 Tongfu Zhonglu Road, Haizhu District, Guangzhou, 510220, Guangdong Province, People's Republic of China,
| | | | | | | | | | | |
Collapse
|
38
|
Galea GL, Meakin LB, Savery D, Taipaleenmaki H, Delisser P, Stein GS, Copp AJ, van Wijnen AJ, Lanyon LE, Price JS. Planar cell polarity aligns osteoblast division in response to substrate strain. J Bone Miner Res 2015; 30:423-35. [PMID: 25264362 PMCID: PMC4333081 DOI: 10.1002/jbmr.2377] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 08/21/2014] [Accepted: 09/10/2014] [Indexed: 02/06/2023]
Abstract
Exposure of bone to dynamic strain increases the rate of division of osteoblasts and also influences the directional organization of the cellular and molecular structure of the bone tissue that they produce. Here, we report that brief exposure to dynamic substrate strain (sufficient to rapidly stimulate cell division) influences the orientation of osteoblastic cell division. The initial proliferative response to strain involves canonical Wnt signaling and can be blocked by sclerostin. However, the strain-related orientation of cell division is independently influenced through the noncanonical Wnt/planar cell polarity (PCP) pathway. Blockade of Rho-associated coiled kinase (ROCK), a component of the PCP pathway, prevents strain-related orientation of division in osteoblast-like Saos-2 cells. Heterozygous loop-tail mutation of the core PCP component van Gogh-like 2 (Vangl2) in mouse osteoblasts impairs the orientation of division in response to strain. Examination of bones from Vangl2 loop-tail heterozygous mice by µCT and scanning electron microscopy reveals altered bone architecture and disorganized bone-forming surfaces. Hence, in addition to the well-accepted role of PCP involvement in response to developmental cues during skeletal morphogenesis, our data reveal that this pathway also acts postnatally, in parallel with canonical Wnt signaling, to transduce biomechanical cues into skeletal adaptive responses. The simultaneous and independent actions of these two pathways appear to influence both the rate and orientation of osteoblast division, thus fine-tuning bone architecture to meet the structural demands of functional loading.
Collapse
Affiliation(s)
- Gabriel L Galea
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Jin H, Wang B, Li J, Xie W, Mao Q, Li S, Dong F, Sun Y, Ke HZ, Babij P, Tong P, Chen D. Anti-DKK1 antibody promotes bone fracture healing through activation of β-catenin signaling. Bone 2015; 71:63-75. [PMID: 25263522 PMCID: PMC4376475 DOI: 10.1016/j.bone.2014.07.039] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 07/11/2014] [Accepted: 07/12/2014] [Indexed: 12/17/2022]
Abstract
In this study we investigated if Wnt/β-catenin signaling in mesenchymal progenitor cells plays a role in bone fracture repair and if DKK1-Ab promotes fracture healing through activation of β-catenin signaling. Unilateral open transverse tibial fractures were created in CD1 mice and in β-catenin(Prx1ER) conditional knockout (KO) and Cre-negative control mice (C57BL/6 background). Bone fracture callus tissues were collected and analyzed by radiography, micro-CT (μCT), histology, biomechanical testing and gene expression analysis. The results demonstrated that treatment with DKK1-Ab promoted bone callus formation and increased mechanical strength during the fracture healing process in CD1 mice. DKK1-Ab enhanced fracture repair by activation of endochondral ossification. The normal rate of bone repair was delayed when the β-catenin gene was conditionally deleted in mesenchymal progenitor cells during the early stages of fracture healing. DKK1-Ab appeared to act through β-catenin signaling to enhance bone repair since the beneficial effect of DKK1-Ab was abrogated in β-catenin(Prx1ER) conditional KO mice. Further understanding of the signaling mechanism of DKK1-Ab in bone formation and bone regeneration may facilitate the clinical translation of this anabolic agent into therapeutic intervention.
Collapse
Affiliation(s)
- Hongting Jin
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical University, Zhejiang, China
| | - Baoli Wang
- Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Jia Li
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA; Liaoning University of Traditional Chinese Medicine, Liaoning, China
| | - Wanqing Xie
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA; Liaoning University of Traditional Chinese Medicine, Liaoning, China
| | - Qiang Mao
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical University, Zhejiang, China
| | - Shan Li
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Fuqiang Dong
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Yan Sun
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical University, Zhejiang, China
| | | | | | - Peijian Tong
- Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical University, Zhejiang, China; Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China.
| | - Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
40
|
Hadjiargyrou M, O'Keefe RJ. The convergence of fracture repair and stem cells: interplay of genes, aging, environmental factors and disease. J Bone Miner Res 2014; 29:2307-22. [PMID: 25264148 PMCID: PMC4455538 DOI: 10.1002/jbmr.2373] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 08/11/2014] [Accepted: 09/10/2014] [Indexed: 01/07/2023]
Abstract
The complexity of fracture repair makes it an ideal process for studying the interplay between the molecular, cellular, tissue, and organ level events involved in tissue regeneration. Additionally, as fracture repair recapitulates many of the processes that occur during embryonic development, investigations of fracture repair provide insights regarding skeletal embryogenesis. Specifically, inflammation, signaling, gene expression, cellular proliferation and differentiation, osteogenesis, chondrogenesis, angiogenesis, and remodeling represent the complex array of interdependent biological events that occur during fracture repair. Here we review studies of bone regeneration in genetically modified mouse models, during aging, following environmental exposure, and in the setting of disease that provide insights regarding the role of multipotent cells and their regulation during fracture repair. Complementary animal models and ongoing scientific discoveries define an increasing number of molecular and cellular targets to reduce the morbidity and complications associated with fracture repair. Last, some new and exciting areas of stem cell research such as the contribution of mitochondria function, limb regeneration signaling, and microRNA (miRNA) posttranscriptional regulation are all likely to further contribute to our understanding of fracture repair as an active branch of regenerative medicine.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | | |
Collapse
|
41
|
Blagodatski A, Poteryaev D, Katanaev VL. Targeting the Wnt pathways for therapies. MOLECULAR AND CELLULAR THERAPIES 2014; 2:28. [PMID: 26056595 PMCID: PMC4452063 DOI: 10.1186/2052-8426-2-28] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 09/05/2014] [Indexed: 12/16/2022]
Abstract
The Wnt/β-catenin signaling pathway is crucial in animal development from sponges to humans. Its activity in the adulthood is less general, with exceptions having huge medical importance. Namely, improper activation of this pathway is carcinogenic in many tissues, most notably in the colon, liver and the breast. On the other hand, the Wnt/β-catenin signaling must be re-activated in cases of tissue damage, and insufficient activation results in regeneration failure and degeneration. These both medically important implications are unified by the emerging importance of this signaling pathway in the control of proliferation of various types of stem cells, crucial for tissue regeneration and, in case of cancer stem cells – cancer progression and relapse. This article aims at briefly reviewing the current state of knowledge in the field of Wnt signaling, followed by a detailed discussion of current medical developments targeting distinct branches of the Wnt pathway for anti-cancer and pro-regeneration therapies.
Collapse
Affiliation(s)
- Artem Blagodatski
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Russian Federation
| | | | - Vladimir L Katanaev
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
42
|
Abstract
OBJECTIVES Wnt5a expression is upregulated during fracture repair and has previously been implicated as a potential regulator of skeletal development and bone mass accrual and maintenance. Our objective was to evaluate the function of Wnt5a in fracture healing. METHODS Femoral fracture experiments on Wnt5a and Wnt5a mice were carried out. To better understand the effect of the Wnt5a on bone repair, we evaluated radiographs using a previously validated qualitative scoring system and performed microcomputed tomography analyses. Histomorphometric analyses determined the temporal distribution of stroma, cartilage matrix, and woven bone in the fracture callus. Finally, we performed tartrate-resistant acid phosphatase (TRAP) immunohistochemical staining to visualize and quantify bone resorbing cells. RESULTS Radiographic evaluations at day 21 demonstrated significantly higher cortical remodeling and bridging parameters for the Wnt5a group compared with the Wnt5a group. The bone volume fraction by microcomputed tomography was also significantly increased in Wnt5a mice. Histological and histomorphometric analyses showed that although Wnt5a mice exhibit decreased cartilage matrix production at day 7 postfracture, they displayed increased residual cartilaginous callus at days 14 and 21 compared with the Wnt5a group. In addition, the total number of multinucleated tartrate-resistant acid phosphatase-positive cells was significantly lower in the Wnt5a group than in the Wnt5a group. CONCLUSIONS The data indicate that decreased Wnt5a signaling impaired proper fracture healing, possibly through decreased cartilaginous callus formation, and delayed cartilage matrix and mineralized tissue remodeling within the fracture callus.
Collapse
|
43
|
Activation of multiple signaling pathways during the differentiation of mesenchymal stem cells cultured in a silicon nanowire microenvironment. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1153-63. [DOI: 10.1016/j.nano.2014.02.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 01/31/2014] [Accepted: 02/13/2014] [Indexed: 12/30/2022]
|
44
|
Liedert A, Röntgen V, Schinke T, Benisch P, Ebert R, Jakob F, Klein-Hitpass L, Lennerz JK, Amling M, Ignatius A. Osteoblast-specific Krm2 overexpression and Lrp5 deficiency have different effects on fracture healing in mice. PLoS One 2014; 9:e103250. [PMID: 25061805 PMCID: PMC4111586 DOI: 10.1371/journal.pone.0103250] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 06/28/2014] [Indexed: 01/09/2023] Open
Abstract
The canonical Wnt/β-catenin pathway plays a key role in the regulation of bone remodeling in mice and humans. Two transmembrane proteins that are involved in decreasing the activity of this pathway by binding to extracellular antagonists, such as Dickkopf 1 (Dkk1), are the low-density lipoprotein receptor related protein 5 (Lrp5) and Kremen 2 (Krm2). Lrp 5 deficiency (Lrp5−/−) as well as osteoblast-specific overexpression of Krm2 in mice (Col1a1-Krm2) result in severe osteoporosis occurring at young age. In this study, we analyzed the influence of Lrp5 deficiency and osteoblast-specific overexpression of Krm2 on fracture healing in mice using flexible and semi-rigid fracture fixation. We demonstrated that fracture healing was highly impaired in both mouse genotypes, but that impairment was more severe in Col1a1-Krm2 than in Lrp5−/− mice and particularly evident in mice in which the more flexible fixation was used. Bone formation was more reduced in Col1a1-Krm2 than in Lrp5−/− mice, whereas osteoclast number was similarly increased in both genotypes in comparison with wild-type mice. Using microarray analysis we identified reduced expression of genes mainly involved in osteogenesis that seemed to be responsible for the observed stronger impairment of healing in Col1a1-Krm2 mice. In line with these findings, we detected decreased expression of sphingomyelin phosphodiesterase 3 (Smpd3) and less active β-catenin in the calli of Col1a1-Krm2 mice. Since Krm2 seems to play a significant role in regulating bone formation during fracture healing, antagonizing KRM2 might be a therapeutic option to improve fracture healing under compromised conditions, such as osteoporosis.
Collapse
Affiliation(s)
- Astrid Liedert
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
- * E-mail:
| | - Viktoria Röntgen
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peggy Benisch
- Orthopaedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Regina Ebert
- Orthopaedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Franz Jakob
- Orthopaedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | | | | | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| |
Collapse
|
45
|
β-Catenin inactivation is a pre-requisite for chick retina regeneration. PLoS One 2014; 9:e101748. [PMID: 25003522 PMCID: PMC4086939 DOI: 10.1371/journal.pone.0101748] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 06/11/2014] [Indexed: 11/19/2022] Open
Abstract
In the present study we explored the role of β-catenin in mediating chick retina regeneration. The chick can regenerate its retina by activating stem/progenitor cells present in the ciliary margin (CM) of the eye or via transdifferentiation of the retinal pigmented epithelium (RPE). Both modes require fibroblast growth factor 2 (FGF2). We observed, by immunohistochemistry, dynamic changes of nuclear β-catenin in the CM and RPE after injury (retinectomy). β-catenin nuclear accumulation was transiently lost in cells of the CM in response to injury alone, while the loss of nuclear β-catenin was maintained as long as FGF2 was present. However, nuclear β-catenin positive cells remained in the RPE in response to injury and were BrdU-/p27+, suggesting that nuclear β-catenin prevents those cells from entering the cell cycle. If FGF2 is present, the RPE undergoes dedifferentiation and proliferation concomitant with loss of nuclear β-catenin. Moreover, retinectomy followed by disruption of active β-catenin by using a signaling inhibitor (XAV939) or over-expressing a dominant negative form of Lef-1 induces regeneration from both the CM and RPE in the absence of FGF2. Our results imply that β-catenin protects cells of the CM and RPE from entering the cell cycle in the developing eye, and specifically for the RPE during injury. Thus inactivation of β-catenin is a pre-requisite for chick retina regeneration.
Collapse
|
46
|
Ellies DL, Economou A, Viviano B, Rey JP, Paine-Saunders S, Krumlauf R, Saunders S. Wise regulates bone deposition through genetic interactions with Lrp5. PLoS One 2014; 9:e96257. [PMID: 24789067 PMCID: PMC4006890 DOI: 10.1371/journal.pone.0096257] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 04/07/2014] [Indexed: 11/18/2022] Open
Abstract
In this study using genetic approaches in mouse we demonstrate that the secreted protein Wise plays essential roles in regulating early bone formation through its ability to modulate Wnt signaling via interactions with the Lrp5 co-receptor. In Wise−/− mutant mice we find an increase in the rate of osteoblast proliferation and a transient increase in bone mineral density. This change in proliferation is dependent upon Lrp5, as Wise;Lrp5 double mutants have normal bone mass. This suggests that Wise serves as a negative modulator of Wnt signaling in active osteoblasts. Wise and the closely related protein Sclerostin (Sost) are expressed in osteoblast cells during temporally distinct early and late phases in a manner consistent with the temporal onset of their respective increased bone density phenotypes. These data suggest that Wise and Sost may have common roles in regulating bone development through their ability to control the balance of Wnt signaling. We find that Wise is also required to potentiate proliferation in chondrocytes, serving as a potential positive modulator of Wnt activity. Our analyses demonstrate that Wise plays a key role in processes that control the number of osteoblasts and chondrocytes during bone homeostasis and provide important insight into mechanisms regulating the Wnt pathway during skeletal development.
Collapse
Affiliation(s)
- Debra L. Ellies
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Androulla Economou
- National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | - Beth Viviano
- Department of Pediatrics, Washington University Medical School, Saint Louis, Missouri, United States of America
| | - Jean-Philippe Rey
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Stephenie Paine-Saunders
- Department of Pediatrics, Washington University Medical School, Saint Louis, Missouri, United States of America
| | - Robb Krumlauf
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
- Department of Anatomy and Cell Biology, Kansas University Medical School, Kansas City, Kansas, United States of America
| | - Scott Saunders
- Department of Pediatrics, Washington University Medical School, Saint Louis, Missouri, United States of America
- Department of Developmental Biology, Washington University Medical School, Saint Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
47
|
Heilmann A, Schinke T, Bindl R, Wehner T, Rapp A, Haffner-Luntzer M, Nemitz C, Liedert A, Amling M, Ignatius A. The Wnt serpentine receptor Frizzled-9 regulates new bone formation in fracture healing. PLoS One 2013; 8:e84232. [PMID: 24391920 PMCID: PMC3877253 DOI: 10.1371/journal.pone.0084232] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 11/21/2013] [Indexed: 12/18/2022] Open
Abstract
Wnt signaling is a key regulator of bone metabolism and fracture healing. The canonical Wnt/β-catenin pathway is regarded as the dominant mechanism, and targeting this pathway has emerged as a promising strategy for the treatment of osteoporosis and poorly healing fractures. In contrast, little is known about the role of non-canonical Wnt signaling in bone. Recently, it was demonstrated that the serpentine receptor Fzd9, a Wnt receptor of the Frizzled family, is essential for osteoblast function and positively regulates bone remodeling via the non-canonical Wnt pathway without involving β-catenin-dependent signaling. Here we investigated whether the Fzd9 receptor is essential for fracture healing using a femur osteotomy model in Fzd9−/− mice. After 10, 24 and 32 days the fracture calli were analyzed using biomechanical testing, histomorphometry, immunohistochemistry, and micro-computed tomography. Our results demonstrated significantly reduced amounts of newly formed bone at all investigated healing time points in the absence of Fzd9 and, accordingly, a decreased mechanical competence of the callus tissue in the late phase of fracture healing. In contrast, cartilage formation and numbers of osteoclasts degrading mineralized matrix were unaltered. β-Catenin immunolocalization showed that canonical Wnt-signaling was not affected in the absence of Fzd9 in osteoblasts as well as in proliferating and mature chondrocytes within the fracture callus. The expression of established differentiation markers was not altered in the absence of Fzd9, whereas chemokines Ccl2 and Cxcl5 seemed to be reduced. Collectively, our results suggest that non-canonical signaling via the Fzd9 receptor positively regulates intramembranous and endochondral bone formation during fracture healing, whereas it does not participate in the formation of cartilage or in the osteoclastic degradation of mineralized matrix. The finding that Fzd9, in addition to its role in physiological bone remodeling, regulates bone repair may have implications for the development of treatments for poorly or non-healing fractures.
Collapse
Affiliation(s)
- Aline Heilmann
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronny Bindl
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Tim Wehner
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Anna Rapp
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Claudia Nemitz
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Astrid Liedert
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
48
|
Ji J, Jia S, Ji K, Jiang WG. Wnt1 inducible signalling pathway protein-2 (WISP‑2/CCN5): roles and regulation in human cancers (review). Oncol Rep 2013; 31:533-9. [PMID: 24337439 DOI: 10.3892/or.2013.2909] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/27/2013] [Indexed: 11/05/2022] Open
Abstract
Wnt1 inducible signalling pathway protein-2 (WISP‑2), also known as CCN5, CT58, CTGF-L, CTGF-3, HICP and Cop1, is one of the 3 WNT1 inducible proteins that belongs to the CCN family. This family of members has been shown to play multiple roles in a number of pathophysiological processes, including cell proliferation, adhesion, wound healing, extracellular matrix regulation, epithelial-mesenchymal transition, angiogenesis, fibrosis, skeletal development and embryo implantation. Recent results suggest that WISP-2 is relevant to tumorigenesis and malignant transformation, particularly in breast cancer, colorectal cancer and hepatocarcinoma. Notably, its roles in cancer appear to vary depending on cell/tumour type and the microenvironment. The striking difference in the structure of WISP-2 in comparison with the other 2 family members may contribute to its difference in functions, which leads to the hypothesis that WISP-2 may act as a dominant-negative regulator of other CCN family members. In the present review, we summarise the roles, regulation and underlying mechanism of WISP-2 in human cancers.
Collapse
Affiliation(s)
- Jiafu Ji
- Department of Gastro-enterological Cancers, Peking University Cancer Hospital, Beijing, P.R. China
| | - Shuqin Jia
- Cardiff University-Peking University Joint Cancer Institute, Beijing, P.R. China
| | - Ke Ji
- Cardiff University-Peking University Joint Cancer Institute, Beijing, P.R. China
| | - Wen G Jiang
- Metastasis and Angiogenesis Research Group, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
49
|
Abstract
STUDY DESIGN To inhibit β-catenin specifically signaling in chondrocytes Col2-ICAT transgenic mice were generated. Anomalies in caudal vertebrae were detected during embryonic and postnatal stages of Col2-ICAT transgenic mice. OBJECTIVE To determine the role of canonical β-catenin signaling in caudal vertebral development. SUMMARY OF BACKGROUND DATA β-catenin signaling plays a critical role in skeletal development. Col2-ICAT transgenic mice were generated to selectively block β-catenin signaling by overexpression of the ICAT gene in chondrocytes. METHODS Tails of E16.5 transgenic embryos and adult Col2-ICAT transgenic mice and their wild-type littermates were collected and analyzed. Skeletal preparation, 3-dimensional micro-computed tomographic and histological analyses were performed to evaluate changes in the structure of caudal vertebrae. Bromodeoxyuridine labeling was performed to evaluate changes in chondrocyte proliferation in caudal vertebrae. RESULTS Skeletal preparation and 3-dimensional micro-computed tomographic analyses revealed bone deformation and angulated deformities in tail tissue in Col2-ICAT transgenic mice. Histological studies revealed abnormal bone development and dysplastic caudal vertebrae in Col2-ICAT transgenic mice. Inhibition of β-catenin signaling in cartilage resulted in vertebral dysplasia leading to aberrant resegmenting process. Thus, 2 poorly developed sclerotomes failed to fuse to form a complete vertebrae. BrdU labeling revealed a decreased chondrocyte proliferation in both cartilageous templates of transgenic embryos and the growth plate of adult Col2-ICAT transgenic mice. CONCLUSION Wnt/β-catenin signaling plays an important role in vertebral development. Inhibition of β-catenin signaling in chondrocytes results in caudal vertebra deformity in mice, which may occur as early as in the stage of sclerotome formation. LEVEL OF EVIDENCE N/A.
Collapse
|
50
|
Yan YB, Li JM, Xiao E, An JG, Gan YH, Zhang Y. A pilot trial on the molecular pathophysiology of traumatic temporomandibular joint bony ankylosis in a sheep model. Part I: Expression of Wnt signaling. J Craniomaxillofac Surg 2013; 42:e15-22. [PMID: 23707780 DOI: 10.1016/j.jcms.2013.04.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 04/11/2013] [Accepted: 04/11/2013] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To preliminarily investigate the temporal patterns of the endogenous mRNA expression for members of the Wnt signaling and a series of genes regulating bone formation during the development of traumatic temporomandibular joint (TMJ) bony ankylosis in a sheep model. METHODS Six sheep were used for the induction of bony ankylosis of TMJ. We performed a condylar fracture, excision of the lateral 2/3 disc and serious injury to the glenoid fossa to induce bony ankylosis on the right TMJ. An isolated condylar fracture was performed on the left side. Two sheep were sacrificed at 1 month, 3 months, and 6 months after surgery, respectively. The specimens from the ankylosed joint and the condylar fracture were harvested for RNA extraction respectively. In this report (Part I), only the bony ankylosed samples were used for analysis of gene expressions. The specimens 1 month postoperatively were taken as the control, and the changes of expression of target genes over time were examined by real-time PCR. RESULTS mRNA expression of Wnt1, Wnt2b, Wnt3a, β-catenin, Sfrp1, Lrp6, Lef1, CyclinD1, and Runx2 was up-regulated at 3 and 6 months compared with 1 month. The expression of Wnt5a, Sox9, and Osterix was up-regulated with a peak at 3 months, and then fell back to the basal levels at 6 months. The expression of Ocn began to up-regulate until 6 month postoperatively. CONCLUSION Our findings suggested that Wnt signaling was involved in the formation of traumatic TMJ bony ankylosis and thus may be a potential therapeutic target for the treatment of the disease in the future.
Collapse
Affiliation(s)
- Ying-Bin Yan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, 22 Zhongguancun Nandajie, Haidian District, Beijing 100081, PR China; Department of Oral and Maxillofacial Surgery, Tianjin Stomatological Hospital, 75 Dagu Road, Heping District, Tianjin 300041, PR China
| | - Jiang-Ming Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, 22 Zhongguancun Nandajie, Haidian District, Beijing 100081, PR China
| | - E Xiao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, 22 Zhongguancun Nandajie, Haidian District, Beijing 100081, PR China
| | - Jin-Gang An
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, 22 Zhongguancun Nandajie, Haidian District, Beijing 100081, PR China
| | - Ye-Hua Gan
- Laboratory of Molecular Biology and Center for Temporomandibular Disorders and Orofacial Pain, Peking University School and Hospital of Stomatology, 22 Zhongguancun Nandajie, Haidian District, Beijing 100081, PR China.
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, 22 Zhongguancun Nandajie, Haidian District, Beijing 100081, PR China.
| |
Collapse
|