1
|
Wada K, Tominaga A, Naruo M, Okazaki K. Managing delayed union of fragility fractures of the pelvis successfully using romosozumab: A case report. Trauma Case Rep 2025; 57:101176. [PMID: 40291407 PMCID: PMC12032325 DOI: 10.1016/j.tcr.2025.101176] [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] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
Fragility fractures of the pelvis (FFPs) are typically caused by minor trauma or without any trauma in older individuals with osteoporosis. In recent years, FFP incidence has increased considerably owing to the increasing number of individuals in the aging population as well as impaired daily life. Surgeries are the main treatment options for some types of FFPs; however, the potential of the use of romosozumab, an FDA-approved humanized monoclonal antibody that can bind and inhibit sclerostin, is yet to be evaluated. Romosozumab substantially increases bone mineral density (BMD) in the spine and the hip, improves bone strength, and prevents the occurrence of new fractures. Previous studies have demonstrated the efficacy of romosozumab in promoting fracture healing, including the healing of nonunion in some fractures. Herein, we present a case of a 61-year-old woman who had FFP delayed union, after falling 4 months before visiting our hospital. She presented with bilateral buttock and leg pain. Baseline BMD measured using dual-energy X-ray absorptiometry revealed a T-score of -3.8 and -3.2 for the lumbar spine and total hip, respectively. As the patient's BMD indicated a high risk of fractures, romosozumab was administered. Her pain improved 3 months after the medication. Computed tomography taken after 3 months revealed that the fracture had healed, suggesting that romosozumab is an effective medication for treating FFP delayed union and nonunion.
Collapse
Affiliation(s)
- K. Wada
- Department of Orthopaedics, Spine Center, Tomei Atsugi Hospital, Kanagawa, Japan
| | - A. Tominaga
- Department of Orthopaedics, Tokyo Women's Medical University, Tokyo, Japan
| | - M. Naruo
- Department of Orthopaedics, Tomei Atsugi Hospital, Kanagawa, Japan
| | - K. Okazaki
- Department of Orthopaedics, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
2
|
Inoue S, Mori M, Yasui M, Matsuki-Fukushima M, Yoshimura K, Nonaka N. Utility of ultrasound imaging in monitoring fracture healing in rat femur: Comparison with other imaging modalities. Bone Rep 2024; 23:101807. [PMID: 39391685 PMCID: PMC11462281 DOI: 10.1016/j.bonr.2024.101807] [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: 07/18/2024] [Revised: 09/19/2024] [Accepted: 09/22/2024] [Indexed: 10/12/2024] Open
Abstract
Objective Fractures are common injuries and various imaging modalities are employed to diagnose and monitor bone union. However, the follow-up of fracture healing using ultrasound imaging (US) remains a topic of debate. In this study, we analyzed of fracture healing process and compared US and radiological analyses with histological analyses to clarify the characteristics and limitations of each modality. Methods An osteotomy model was created using the femur of Wistar rats, and US, radiological (radiography and micro-computed tomography (micro-CT)), and histological analyses were performed. Radiological assessments were conducted for the evaluation of calcified tissue. The gap between the bony callus and cartilaginous callus was measured. Results US effectively captured changes on the fracture surface, potentially reflecting the early healing processes. Both US and radiographic findings showed strong correlation in terms of the decrease in the bony callus gap. US was unable to distinguish cartilaginous callus from the surrounding soft tissue. During the remodeling stage, micro-CT offered a detailed assessment of the internal fracture surface, whereas US was limited to evaluating the outer bone surface and lacked accuracy in visualizing the entire fracture site. Radiography provided a general overview of the fractures. The decrease in the bony callus gap measured using US correlated with the reduction in cartilaginous callus observed histologically. Conclusion This study demonstrated that US could be a valuable tool for evaluating fracture healing. Combining fracture management with US and radiological examinations may provide a more accurate assessment of healing progress.
Collapse
Affiliation(s)
- Satoshi Inoue
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Michinori Mori
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
- Department of Judo Physical Therapy, Faculty of Health Care, Teikyo Heisei University, 2-51-4 Higashi-Ikebukuro, Toshimaku, Tokyo, Japan
| | - Masaya Yasui
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
- Department of Judo Seifuku and Health Sciences, Tokoha University, 1230 Miyakoda-Cho, Kita-Ku, Hamamatsu, Shizuoka 431-2102, Japan
| | - Miwako Matsuki-Fukushima
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Kentaro Yoshimura
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Naoko Nonaka
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| |
Collapse
|
3
|
Tachibana S, Hayashi S, Ikuta K, Anjiki K, Onoi Y, Suda Y, Wada K, Maeda T, Saito A, Tsubosaka M, Kamenaga T, Kuroda Y, Nakano N, Matsumoto T, Hosooka T, Ogawa W, Kuroda R. Downregulation of Krüppel-like factor 15 expression delays endochondral bone ossification during fracture healing. Bone 2024; 190:117302. [PMID: 39437873 DOI: 10.1016/j.bone.2024.117302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE The role of Krüppel-like zinc finger transcription factor 15 (KLF15) in endochondral ossification during fracture healing remains unexplored. In this study, we aimed to elucidate the impact of KLF15 in a mouse model of tibial transverse fracture. METHODS We created tamoxifen-inducible, cartilage-specific KLF15 knockout mice (KLF15 KO). KLF15 fl/fl Col2-CreERT mice from the same litters as the KLF15 KO mice, but not treated with 4-hydroxytamoxifen, were used as controls (CT). At 10 weeks, male KLF15 KO and CT mice underwent tibial fracture followed by intramedullary nailing. Both groups were administered tamoxifen at days 0, 3, and 7 after surgery. The tibiae were harvested on post-surgery days 7, 10, and 14 for radiological assessment using micro-computed tomography. Histological staining (Safranin-O) and immunohistochemistry for KLF15, SOX9, Indian hedgehog (IHH), RUNX2, and Osterix were performed. Additionally, cartilage from mouse fetus was cultured for qRT-PCR and western blot analyses of KLF15, SOX9, IHH, Col2, RUNX2, Osterix, TGF-β, SMAD3, and phosphor-SMAD3. RESULTS The radiological assessment revealed that immature callus formation was delayed in KLF15 KO, compared with that in CT, peaking on day 14 compared with that on day 10 in CT. KLF15 KO mice exhibited delayed fracture healing and reduced Safranin-O staining at days 7 and 10 post-surgery. The ratio of cells positive for KLF15 and SOX9 was significantly lower in KLF15 KO than in CT, whereas the ratios for IHH, RUNX2, and Osterix showed no significant difference. RT-PCR revealed reduced expression of KLF15, SOX9, and COL2, with no significant changes in IHH, Osterix, RUNX2, TGF-β, and SMAD3. Western blot analysis indicated decreased SMAD3 phosphorylation in KLF15 KO mice. CONCLUSION KLF15 regulates SOX9 via the TGF-β-SMAD3-SOX9 pathway, independent of IHH, in endochondral ossification. The KLF15 deficiency decreases SOX9 expression through reduced SMAD3 phosphorylation, subsequently delaying fracture healing.
Collapse
Affiliation(s)
- Shotaro Tachibana
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Kemmei Ikuta
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kensuke Anjiki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuma Onoi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshihito Suda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kensuke Wada
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takuma Maeda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akira Saito
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoki Nakano
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tetsuya Hosooka
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
4
|
Wong RMY, Wong PY, Liu C, Wong HY, Fong MK, Zhang N, Cheung WH, Law SW. Treatment effects, adverse outcomes and cardiovascular safety of romosozumab - Existing worldwide data: A systematic review and meta-analysis. J Orthop Translat 2024; 48:107-122. [PMID: 39189010 PMCID: PMC11345130 DOI: 10.1016/j.jot.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Background Romosozumab is a novel monoclonal antibody that binds to sclerostin, and has dual effects of increasing bone formation and decreasing bone resorption, giving it a unique mechanism of action. The objective of this study was to perform a systematic review and meta-analysis based on existing worldwide data on treatment effects and safety of romosozumab in randomized controlled trials. Methods A systematic search was carried out on four databases including PubMed, Embase, Web of Science and Cochrane Central Register of Controlled Trials (CENTRAL). The keywords used for search was "(romosozumab) AND (osteoporosis OR safety)". Randomized controlled trial or post-hoc studies of the included randomized controlled trial which studied the effects and safety of romosozumab were included. The quality of selected studies was assessed with the Cochrane collaboration tool and the PEDro scale. Results 20 studies were included for qualitative analysis. 14 studies were included for meta-analysis. In total, there were 13,507 (n = 13,507) participants with 637 men and 12,870 women from original cohorts. The overall mean difference was in favor of romosozumab treatment for lumbar spine (10.04 (95 % confidence interval (CI) = 7.51-12.57; p < 0.00001)), total hip (4.04 (95 % CI = 3.10-4.99; p < 0.00001)) and femoral neck bone mineral density (3.77 (95 % CI = 2.90-4.64; p < 0.00001)) at 12 months. There was significantly less likelihood of new vertebral fractures with romosozumab compared to control (odds ratio (OR) 0.42 (95 % CI = 0.20-0.89); p = 0.02) at 12 months of treatment. There was significantly less likelihood of new vertebral fracture at 24 months with 12 months of romosozumab followed by sequential treatment with anti-resorptive compared to control with only anti-resorptive agent use (OR 0.36 (95 % CI = 0.18-0.71); p = 0.003). There was no significant difference in serious adverse events and fatal adverse events with use of romosozumab compared with control in our meta-analyses. There were no significant differences in serious cardiovascular events in Asian population of romosozumab with control group with 12 months of romosozumab treatment followed by 24 months of anti-resorptive agent with OR 1.09 (95 % CI = 0.40-2.96; P = 0.86). There was no significant difference between romosozumab group and control group for the median time to radiographic healing. Our qualitative analysis on Quantitative Computed Tomography (QCT), Finite element analysis (FEA) and bone biopsy analyses demonstrated that romosozumab improved parameters and measures in these domains as well. Conclusion In conclusion, our study showed that romosozumab was an effective agent to treat osteoporosis with high quality evidence. There were no significant differences in the adverse events, serious adverse events, fatal adverse events identified. Further subgroup analysis of cardiovascular events and cardiovascular death in the total population showed no differences either. The translational potential of this article Given the results, romosozumab is an effective agent to treat patients with very-high risk of osteoporotic fractures.
Collapse
Affiliation(s)
- Ronald Man Yeung Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Pui Yan Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chaoran Liu
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hei Yuet Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Man Ki Fong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ning Zhang
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing Hoi Cheung
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sheung Wai Law
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
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
|
6
|
Zhang B, Swanson WB, Durdan M, Livingston HN, Dodd M, Vidanapathirana SM, Desai A, Douglas L, Mishina Y, Weivoda M, Greineder CF. Affinity targeting of therapeutic proteins to the bone surface-local delivery of sclerostin-neutralizing antibody enhances efficacy. J Bone Miner Res 2024; 39:717-728. [PMID: 38526976 PMCID: PMC11472147 DOI: 10.1093/jbmr/zjae050] [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: 11/30/2023] [Revised: 03/02/2024] [Accepted: 03/23/2024] [Indexed: 03/27/2024]
Abstract
Currently available biotherapeutics for the treatment of osteoporosis lack explicit mechanisms for bone localization, potentially limiting efficacy and inducing off-target toxicities. While various strategies have been explored for targeting the bone surface, critical aspects remain poorly understood, including the optimal affinity ligand, the role of binding avidity and circulation time, and, most importantly, whether or not this strategy can enhance the functional activity of clinically relevant protein therapeutics. To investigate, we generated fluorescent proteins (eg, mCherry) with site-specifically attached small molecule (bisphosphonate) or peptide (deca-aspartate, D10) affinity ligands. While both affinity ligands successfully anchored fluorescent protein to the bone surface, quantitative radiotracing revealed only modest femoral and vertebral accumulation and suggested a need for enhanced circulation time. To achieve this, we fused mCherry to the Fc fragment of human IgG1 and attached D10 peptides to each C-terminus. The mCherry-Fc-D10 demonstrated an ~80-fold increase in plasma exposure and marked increases in femoral and vertebral accumulation (13.6% ± 1.4% and 11.4% ± 1.3% of the injected dose/g [%ID/g] at 24 h, respectively). To determine if bone surface targeting could enhance the efficacy of a clinically relevant therapeutic, we generated a bone-targeted sclerostin-neutralizing antibody, anti-sclerostin-D10. The targeted antibody demonstrated marked increases in bone accumulation and retention (20.9 ± 2.5% and 19.5 ± 2.5% ID/g in femur and vertebrae at 7 days) and enhanced effects in a murine model of ovariectomy-induced bone loss (bone volume/total volume, connectivity density, and structure model index all increased [P < .001] vs untargeted anti-sclerostin). Collectively, our results indicate the importance of both bone affinity and circulation time in achieving robust targeting of therapeutic proteins to the bone surface and suggest that this approach may enable lower doses and/or longer dosing intervals without reduction in biotherapeutic efficacy. Future studies will be needed to determine the translational potential of this strategy and its potential impact on off-site toxicities.
Collapse
Affiliation(s)
- Boya Zhang
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - William Benton Swanson
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margaret Durdan
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Hematology, Mayo Clinic, Rochester, MN 55905, USA
| | - Heather N Livingston
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michaela Dodd
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sachith M Vidanapathirana
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alec Desai
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lindsey Douglas
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yuji Mishina
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Megan Weivoda
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Hematology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Colin F Greineder
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Emergency Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Ozawa Y, Takegami Y, Osawa Y, Asamoto T, Tanaka S, Imagama S. Anti-sclerostin antibody therapy prevents post-ischemic osteonecrosis bone collapse via interleukin-6 association. Bone 2024; 181:117030. [PMID: 38309414 DOI: 10.1016/j.bone.2024.117030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Osteonecrosis of the femoral head (ONFH) is a debilitating condition characterized by subchondral bone necrosis, which frequently culminates in joint destruction. Although total hip arthroplasty is conventionally practiced to remediate ONFH, for patients under the age of 60, the outcomes can be suboptimal. Chronic inflammation, particularly that mediated by interleukin-6 (IL-6), has been conjectured to be a potential mechanism underlying the etiology of ONFH. This study aimed at exploring the interplay between IL-6, the canonical Wnt signaling pathway, and ONFH to provide insights for potential therapeutic interventions. Human ONFH specimens depicted an elevation in β-catenin expression in the transitional layer, while IL-6 levels were pronounced in the same region. Subsequently, mouse models of ischemic osteonecrosis were treated with an anti-sclerostin antibody to assess its effects on bone metabolism and cellular processes. Histological analysis revealed that the administration of anti-sclerostin antibodies effectuated early recovery from bone necrosis, reduced empty lacunae, and suppressed IL-6 expression. The treatment evidently initiated the activation of the Wnt/β-catenin signaling pathway, presenting a potential mechanism associated with IL-6-mediated inflammation. Furthermore, the antibody upregulated osteoblast formation, downregulated osteoclast formation, and increased bone volume. Micro-CT imaging demonstrated increased bone volume, prevented epiphyseal deformity, and improved compression strength. Therefore, this study yields significant findings, indicating the potency of anti-sclerostin antibodies in effectively modulating the Wnt/β-catenin pathway, associating with IL-6 expression, and preventing post-ONFH bone collapse. Additionally, this preclinical investigation in mouse models offers an avenue for prospective research on potential therapeutic interventions against human ONFH.
Collapse
Affiliation(s)
- Yuto Ozawa
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Yasuhiko Takegami
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan.
| | - Yusuke Osawa
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Takamune Asamoto
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Shinya Tanaka
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Shiro Imagama
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| |
Collapse
|
8
|
Suen PK, Zheng L, Yang QQ, Mak WS, Pak WY, Mo KY, Chan ML, Liu QQ, Qin L, Sun SSM. Lysine-rich rice partially enhanced the growth and development of skeletal system with better skeletal microarchitecture in young rats. Nutr Res 2024; 121:67-81. [PMID: 38043437 DOI: 10.1016/j.nutres.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 12/05/2023]
Abstract
Rice is the primary staple food for half of the world's population but is low in lysine content. Previously, we developed transgenic rice with enhanced free lysine content in rice seeds (lysine-rich rice), which was shown safe for consumption and improved the growth in rats. However, the effects of lysine-rich rice on skeletal growth and development remained unknown. In this study, we hypothesized that lysine-rich rice improved skeletal growth and development in weaning rats. Male weaning Sprague-Dawley rats received lysine-rich rice (HFL) diet, wild-type rice (WT) diet, or wild-type rice with various contents of lysine supplementation diet for 70 days. Bone microarchitectures were examined by microcomputed tomography, bone strength was investigated by mechanical test, and dynamics of bone growth were examined by histomorphometric analysis. In addition, we explored the molecular mechanism of lysine and skeletal growth through biochemical testing of growth hormone, bone turnover marker, and amino acid content of rat serum analysis, as well as in a cell culture system. Results indicated that the HFL diet improved rats' bone growth, strength, and microarchitecture compared with the WT diet group. In addition, the HFL diet increased the serum essential amino acids, growth hormone (insulin-like growth factor-1), and bone formation marker concentrations. The cell culture model showed that lysine deficiency reduced insulin-like growth factor-1 and Osterix expression, Akt/mammalian target of rapamycin signaling, and matrix mineralization, and inhibited osteoblast differentiation associated with bone growth. Our findings showed that lysine-rich rice improved skeletal growth and development in weaning rats. A further increase of rice lysine content is highly desirable to fully optimize bone growth and development.
Collapse
Affiliation(s)
- Pui Kit Suen
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China; Institute of Plant Molecular Biology and Agriculture Biotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Science, China
| | - Qing-Qing Yang
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China; China Key Laboratory of Plant Functional Genomics of the Ministry of Education, College of Agriculture, Yangzhou University, Yangzhou, China
| | - Wan Sheung Mak
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wan Yu Pak
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kit Ying Mo
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Man-Ling Chan
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qiao-Quan Liu
- China Key Laboratory of Plant Functional Genomics of the Ministry of Education, College of Agriculture, Yangzhou University, Yangzhou, China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Samuel Sai-Ming Sun
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China; Institute of Plant Molecular Biology and Agriculture Biotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
9
|
Sheng R, Cao M, Song M, Wang M, Zhang Y, Shi L, Xie T, Li Y, Wang J, Rui Y. Muscle-bone crosstalk via endocrine signals and potential targets for osteosarcopenia-related fracture. J Orthop Translat 2023; 43:36-46. [PMID: 38021216 PMCID: PMC10654153 DOI: 10.1016/j.jot.2023.09.007] [Citation(s) in RCA: 7] [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: 02/06/2023] [Revised: 08/14/2023] [Accepted: 09/20/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Osteosarcopenia is a syndrome coexisting sarcopenia and osteopenia/osteoporosis, with a high fracture risk. Recently, skeletal muscle and bone have been recognized as endocrine organs capable of communication through secreting myokines and osteokines, respectively. With a deeper understanding of the muscle-bone crosstalk, these endocrine signals exhibit an important role in osteosarcopenia development and fracture healing. METHODS This review summarizes the role of myokines and osteokines in the development and treatment of osteosarcopenia and fracture, and discusses their potential for osteosarcopenia-related fracture treatment. RESULTS Several well-defined myokines (myostatin and irisin) and osteokines (RANKL and SOST) are found to not only regulate skeletal muscle and bone metabolism but also influence fracture healing processes. Systemic interventions targeting these biochemical signals has shown promising results in improving the mass and functions of skeletal muscle and bone, as well as accelerating fracture healing processes. CONCLUSION The regulation of muscle-bone crosstalk via biochemical signals presents a novel and promising strategy for treating osteosarcopenia and fracture by simultaneously enhancing bone and muscle anabolism. We propose that myostatin, irisin, RANKL, and SOST may serve as potential targets to treat fracture patients with osteosarcopenia. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Osteosarcopenia is an emerging geriatric syndrome where sarcopenia and osteoporosis coexist, with high fracture risk, delayed fracture healing, and increased mortality. However, no pharmacological agent is available to treat fracture patients with osteosarcopenia. This review summarizes the role of several myokines and osteokines in the development and treatment of osteosacropenia and fracture, as well as discusses their potential as intervention targets for osteosarcopenia-related fracture, which provides a novel and promising strategy for future osteosarcopenia-related fracture treatment.
Collapse
Affiliation(s)
- Renwang Sheng
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Mumin Cao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Mingyuan Song
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Mingyue Wang
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Yuanwei Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Liu Shi
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Tian Xie
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Yingjuan Li
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Department of Geriatrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Jinyu Wang
- Department of Rehabilitation, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
- Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| |
Collapse
|
10
|
Sangadala S, Kim CH, Fernandes LM, Makkar P, Beck GR, Boden SD, Drissi H, Presciutti SM. Sclerostin small-molecule inhibitors promote osteogenesis by activating canonical Wnt and BMP pathways. eLife 2023; 12:e63402. [PMID: 37560905 PMCID: PMC10431921 DOI: 10.7554/elife.63402] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 08/09/2023] [Indexed: 08/11/2023] Open
Abstract
Background The clinical healing environment after a posterior spinal arthrodesis surgery is one of the most clinically challenging bone-healing environments across all orthopedic interventions due to the absence of a contained space and the need to form de novo bone. Our group has previously reported that sclerostin in expressed locally at high levels throughout a developing spinal fusion. However, the role of sclerostin in controlling bone fusion remains to be established. Methods We computationally identified two FDA-approved drugs, as well as a single novel small-molecule drug, for their ability to disrupt the interaction between sclerostin and its receptor, LRP5/6. The drugs were tested in several in vitro biochemical assays using murine MC3T3 and MSCs, assessing their ability to (1) enhance canonical Wnt signaling, (2) promote the accumulation of the active (non-phosphorylated) form of β-catenin, and (3) enhance the intensity and signaling duration of BMP signaling. These drugs were then tested subcutaneously in rats as standalone osteoinductive agents on plain collagen sponges. Finally, the top drug candidates (called VA1 and C07) were tested in a rabbit posterolateral spine fusion model for their ability to achieve a successful fusion at 6 wk. Results We show that by controlling GSK3b phosphorylation our three small-molecule inhibitors (SMIs) simultaneously enhance canonical Wnt signaling and potentiate canonical BMP signaling intensity and duration. We also demonstrate that the SMIs produce dose-dependent ectopic mineralization in vivo in rats as well as significantly increase posterolateral spine fusion rates in rabbits in vivo, both as standalone osteogenic drugs and in combination with autologous iliac crest bone graft. Conclusions Few if any osteogenic small molecules possess the osteoinductive potency of BMP itself - that is, the ability to form de novo ectopic bone as a standalone agent. Herein, we describe two such SMIs that have this unique ability and were shown to induce de novo bone in a stringent in vivo environment. These SMIs may have the potential to be used in novel, cost-effective bone graft substitutes for either achieving spinal fusion or in the healing of critical-sized fracture defects. Funding This work was supported by a Veteran Affairs Career Development Award (IK2-BX003845).
Collapse
Affiliation(s)
- Sreedhara Sangadala
- Atlanta Veterans Affairs Medical CenterDecaturUnited States
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
| | - Chi Heon Kim
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
| | - Lorenzo M Fernandes
- Atlanta Veterans Affairs Medical CenterDecaturUnited States
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
| | - Pooja Makkar
- Department of Biotechnology, Panjab UniversityChandigarhIndia
| | - George R Beck
- Atlanta Veterans Affairs Medical CenterDecaturUnited States
- Emory University, Division of EndocrinologyAtlantaUnited States
| | - Scott D Boden
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
| | - Hicham Drissi
- Atlanta Veterans Affairs Medical CenterDecaturUnited States
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
| | - Steven M Presciutti
- Atlanta Veterans Affairs Medical CenterDecaturUnited States
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
| |
Collapse
|
11
|
Florio M, Kostenuik PJ, Stolina M, Asuncion FJ, Grisanti M, Ke HZ, Ominsky MS. Dual Inhibition of the Wnt Inhibitors DKK1 and Sclerostin Promotes Fracture Healing and Increases the Density and Strength of Uninjured Bone: An Experimental Study in Nonhuman Primates. J Bone Joint Surg Am 2023; 105:1145-1155. [PMID: 37159527 DOI: 10.2106/jbjs.22.01092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
BACKGROUND Fracture repair involves the reactivation of developmental signaling cascades, including Wnt signaling that stimulates bone formation and bone regeneration. Rodent data indicate that dual inhibition of the Wnt signaling antagonists sclerostin and Dickkopf-1 (DKK1) increases callus bone volume and strength while increasing bone mass systemically. METHODS We evaluated the effects of 16 weeks of subcutaneously administered carrier solution (vehicle, VEH), anti-sclerostin antibody (Scl-Ab), anti-DKK1 antibody (DKK1-Ab), or Scl-Ab plus DKK1-Ab combination therapy (COMBO) on ulnar osteotomy healing in nonhuman primates (cynomolgus monkeys; 20 to 22 per group). RESULTS Scl-Ab and COMBO therapy increased systemic markers of bone formation versus VEH, with COMBO leading to synergistic increases versus Scl-Ab or DKK1-Ab monotherapies. The COMBO and Scl-Ab groups showed reduced serum markers of bone resorption versus VEH. The COMBO and DKK1-Ab groups exhibited greater callus bone mineral density (BMD), torsional stiffness, and torsional rigidity versus VEH. Lumbar vertebrae from the Scl-Ab and COMBO groups showed greater BMD and bone formation rate versus VEH, and the femoral mid-diaphysis of the Scl-Ab and COMBO groups showed greater periosteal and endocortical bone formation rates versus VEH. CONCLUSIONS DKK1-Ab increased BMD and strength at the ulnar osteotomy site, Scl-Ab increased bone formation and BMD at uninjured skeletal sites, and Scl-Ab plus DKK1-Ab combination therapy induced all of these effects, in some cases to a greater degree versus 1 or both monotherapies. These results in nonhuman primates suggest that DKK1 preferentially regulates bone healing while sclerostin preferentially regulates systemic bone mass. CLINICAL RELEVANCE Combination therapy with antibodies against sclerostin and DKK1 may offer a promising therapeutic strategy for both fracture treatment and fracture prevention.
Collapse
Affiliation(s)
- Monica Florio
- Discovery Research, Amgen, Thousand Oaks, California
| | - Paul J Kostenuik
- Discovery Research, Amgen, Thousand Oaks, California
- Phylon Pharma Services, Thousand Oaks, California
- University of Michigan School of Dentistry, Ann Arbor, Michigan
| | | | | | | | - Hua Zhu Ke
- Discovery Research, Amgen, Thousand Oaks, California
- Angitia Biopharmaceuticals, Guangzhou, Guangdong, People's Republic of China
| | - Michael S Ominsky
- Discovery Research, Amgen, Thousand Oaks, California
- Ascendis Pharma, Palo Alto, California
| |
Collapse
|
12
|
Nurrachman AS, Azhari A, Epsilawati L, Pramanik F. Temporal Pattern of micro-CT Angiography Vascular Parameters and VEGF mRNA Expression in Fracture Healing: a Radiograph and Molecular Comparison. Eur J Dent 2023. [PMID: 36716788 DOI: 10.1055/s-0042-1757466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis plays an important role in fracture healing with vascular endothelial growth factor (VEGF) as the main protein involved. Micro-computed tomography (CT) angiography may be used to analyze this revascularization with several parameters such as number of branches, total volume, and diameter. This systematic review is aimed to assess available studies on the temporal pattern of vascular imaging on micro-CT angiographs, especially in terms of the number of branches, total volume, and diameter as well as the temporal pattern of VEGF mRNA expression as the molecular comparison during bone fracture healing. This review was conducted according to the PRISMA guidelines. Electronic database searches were performed using PubMed, ProQuest, ScienceDirect, EBSCOhost, Taylor & Francis Online, and hand searching. The search strategy and keywords were adjusted to each database using the Boolean operators and other available limit functions to identify most relevant articles based on our inclusion and exclusion criteria. Screening and filtration were done in several stages by removing the duplicates and analyzing each title, abstract, and full-text in all included entries. Data extraction was done for syntheses to summarize the temporal pattern of each parameter. A total of 28 articles were eligible and met all criteria, 11 articles were synthesized in its angiograph's analysis, 16 articles were synthesized in its VEGF mRNA expression analysis, and 1 article had both parameters analyzed. The overall temporal pattern of both three micro-CT angiographic parameters and VEGF mRNA expression was in line qualitatively. The number of branches, total volume, and diameter of the blood vessels in micro-CT angiography showed an exponential rise at week 2 and decline at week 3 of fracture healing, with the VEGF mRNA expression concurrently showing a consistent pattern in the phase.
Collapse
Affiliation(s)
- Aga Satria Nurrachman
- Department of Oral and Maxillofacial Radiology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Azhari Azhari
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, West Java, Indonesia
| | - Lusi Epsilawati
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, West Java, Indonesia
| | - Farina Pramanik
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, West Java, Indonesia
| |
Collapse
|
13
|
Jin Y. Analysis of radiologic and clinical outcome in acute osteoporotic vertebral compression fracture: Single-agent teriparatide vs. teriparatide with subsequent vertebroplasty. World Neurosurg X 2023; 18:100153. [PMID: 36818733 PMCID: PMC9932136 DOI: 10.1016/j.wnsx.2023.100153] [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: 09/01/2022] [Accepted: 01/10/2023] [Indexed: 01/22/2023] Open
Abstract
Objectives To analyze the difference in union and clinical outcomes between teriparatide (T) and teriparatide with vertebroplasty (V) treatment modalities in osteoporotic vertebral compression fractures (OVCFs). Methods Patients were divided into two groups (T and V: 87 and 92 patients with 105 fractures each). Radiological features (fracture type/grade, presence of fracture gap/intravertebral vacuum cleft (IVVC)/posterior vertebral wall fracture, change in compression rate (CR)/kyphotic angle (CA), and fusion status) were assessed with 3D-CT at 3 and 6 months. The outcome was divided into success or failure based on visual analog scale (<3), absence of percussion tenderness on the spinous process, and pain during motion. Univariate and multivariate analyses were performed to identify risk factors for nonunion and failed outcomes in each group. Results The V group showed more favorable results than the T group at 3 months (CR>10%, 58% vs. 17%; CA>5°, 36% vs. 16%; union, 66% vs. 91%; successful outcome, 77% vs. 94%). At 6 months, no significant change was detected in CR and CA. A significant difference remained in union (89% vs. 100%) and successful outcomes (79% vs. 100%). The V group with age (>75 years) and initial CR (>40%) had more benefits than the T group in the subgroup analysis. In multivariate analysis for the T group, nonunion risk factors were hypertension (P = .0054) and fracture gap (P = .0075). IVVC (P = .047) was the sole risk factor for failure. Conclusions Teriparatide with subsequent vertebroplasty can be selected as the first-line treatment with better sequelae and outcomes in acute osteoporotic compression fractures.
Collapse
Affiliation(s)
- Yongjun Jin
- Department of Neurosurgery, Seoul Paik Hospital, Inje University College of Medicine 04551, Mareunnae-ro 9, Jung-gu, Seoul, South Korea.
| |
Collapse
|
14
|
Kikuchi K, Haneda M, Hayashi S, Maeda T, Nakano N, Kuroda Y, Tsubosaka M, Kamenaga T, Fujita M, Ikuta K, Anjiki K, Tachibana S, Onoi Y, Matsumoto T, Kuroda R. P21 deficiency exhibits delayed endochondral ossification during fracture healing. Bone 2022; 165:116572. [PMID: 36180020 DOI: 10.1016/j.bone.2022.116572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Endochondral ossification is a complex biological phenomenon involving a variety of factors and cells. Cyclin-dependent kinase inhibitor 1 (p21) inhibits cell cycle progression and is affected by external stress. We recently reported that embryonic endochondral ossification is unaffected by endogenous p21 deficiency. In this study, we evaluated whether p21 expression affects endochondral ossification during fracture healing. METHODS Tibial fractures were introduced into p21 knockout (p21-/-) (n = 24) and wild-type C57BL/6 (p21+/+) (n = 24) mice at age 10 weeks. Fracture healing was evaluated using radiological, histological, and immunohistochemical (IHC) analyses. The effect of p21 small interfering RNA (siRNA) on ATDC5 cells was assessed in vitro. RESULTS The Allen score for fracture healing was lower in p21-/- mice than in p21+/+ mice. In addition, p21-/- mice exhibited larger calluses and lower bone mineral density. IHC analyses showed that p21-/- mice exhibited delayed endochondral ossification via the Ihh-Runx2-Osterix pathway in vivo. Down-regulation of p21 expression in ATDC5 cells delayed endochondral ossification in vitro. CONCLUSIONS p21 deficiency leads to delayed endochondral ossification by attenuating the Ihh-Runx2-Osterix pathway in vivo, and p21 deficiency in hypertrophic chondrocytes causes delayed differentiation of hypertrophic chondrocytes in vitro. p21 plays a role in endochondral ossification during fracture healing.
Collapse
Affiliation(s)
- Kenichi Kikuchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahiko Haneda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Toshihisa Maeda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoki Nakano
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahiro Fujita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenmei Ikuta
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kensuke Anjiki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shotaro Tachibana
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuma Onoi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
15
|
Kan T, He Z, Du J, Xu M, Cui J, Han X, Tong D, Li H, Yan M, Yu Z. Irisin promotes fracture healing by improving osteogenesis and angiogenesis. J Orthop Translat 2022; 37:37-45. [PMID: 36196152 PMCID: PMC9513699 DOI: 10.1016/j.jot.2022.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 06/27/2022] [Accepted: 07/15/2022] [Indexed: 11/08/2022] Open
Abstract
Background Osteogenesis and angiogenesis are important for bone fracture healing. Irisin is a muscle-derived monokine that is associated with bone formation. Methods To demonstrate the effect of irisin on bone fracture healing, closed mid-diaphyseal femur fractures were produced in 8-week-old C57BL/6 mice. Irisin was administrated intraperitoneally every other day after surgery, fracture healing was assessed by using X-rays. Bone morphometry of the fracture callus were assessed by using micro-computed tomography. Femurs of mice from each group were assessed by the three-point bending testing. Effect of irisin on osteogenic differentiation in mesenchymal stem cells in vitro was evaluated by quantitative real-time polymerase chain reaction (qRT-PCR), alkaline phosphatase staining and alizarin red staining. Angiogenesis of human umbilical vein endothelial cells (HUVECs) were evaluated by qRT-PCR, migration tests, and tube formation assays. Results Increased callus formation, mineralization and tougher fracture healing were observed in the irisin-treated group than in the control group, indicating the better fracture callus healing due to Irisin treatment. The vessel surface and vessel volume fraction of the callus also increased in the irisin-treated group. The expression of BMP2, CD31, and VEGF in callus were enhanced in the irisin-treated group. In mouse bone mesenchymal stem cells, irisin promoted ALP expression and mineralization, and increased the expression of osteogenic genes, including OSX, Runx2, OPG, ALP, OCN and BMP2. Irisin also promoted HUVEC migration and tube formation. Expression of angiogenic genes, including ANGPT1, ANGPT2, VEGFb, CD31, FGF2, and PDGFRB in HUVECs were increased by irisin. Conclusion All the results indicate irisin can promote fracture healing through osteogenesis and angiogenesis. These findings help in the understanding of muscle–bone interactions during fracture healing. The Translational Potential of this Article Irisin was one of the most important monokine secreted by skeletal muscle. Studies have found that irisin have anabolic effect one bone remodeling through affecting osteocyte and osteoblast. Based on our study, irisin could promote bone fracture healing by increasing bone mass and vascularization, which provide a potential usage of irisin to promote fracture healing and improve clinical outcomes.
Collapse
|
16
|
Takase R, Tsubouchi Y, Otsu T, Kataoka T, Iwasaki T, Kataoka M, Tsumura H. The effects of romosozumab combined with active vitamin D 3 on fracture healing in ovariectomized rats. J Orthop Surg Res 2022; 17:384. [PMID: 35962437 PMCID: PMC9373334 DOI: 10.1186/s13018-022-03276-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/02/2022] [Indexed: 11/10/2022] Open
Abstract
Background In this study, we investigated the potential acceleration of fracture healing and bone mineral density-increasing effects of romosozumab and active vitamin D3 combination therapy for fractures in ovariectomized rats. Methods Ovariectomy was performed on 40 24-week-old female Sprague–Dawley rats. After 8 weeks, the rats were subjected to periosteum removal and osteotomy of the femoral shaft followed by osteosynthesis with intramedullary nailing to create fracture models. The rats were then divided into four groups: C group (control), R group (receiving romosozumab at 25 mg/kg once a month via subcutaneous injection), VD group (receiving active vitamin D3 at 0.2 µg/kg twice a week via subcutaneous injection), and R + VD group. Further, 10 rats were included in a sham group. At 10 weeks after the intervention, both femurs were removed and blood samples were collected from all rats. Soft X-ray imaging was used to evaluate bone union, and microcomputed tomography (micro-CT) was used for bone morphometric evaluation. Toluidine blue staining was used for the histopathological evaluation of the undecalcified specimens, and bone turnover marker levels were measured using enzyme-linked immunosorbent assay. Results Bone morphometry analysis via micro-CT revealed increased mineral density of the trabecular bone in the R + VD group femurs, demonstrating the effectiveness of romosozumab plus active vitamin D3 combination therapy. However, there were no differences in bone union evaluated using soft X-ray imaging, indicating no acceleration of fracture healing. Conclusions Although romosozumab and active vitamin D3 combination therapy increased trabecular bone volume, there was no evidence on its ability to accelerate fracture healing. Graphical abstract ![]()
Collapse
Affiliation(s)
- Ryota Takase
- Oita University Hospital Rehabilitation Center, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu-city, Oita, 879-5593, Japan
| | - Yuta Tsubouchi
- School of Physical Therapy, Faculty of Rehabilitation, Reiwa Health Sciences University, 2-1-12 Wajirogaoka, Higashi-ku, Fukuoka, 811-0213, Japan
| | - Takefumi Otsu
- Division of Mechatronics, Department of Innovative Engineering, Faculty of Science and Technology, Oita University, 700 Dannoharu, Oita, 870-1192, Japan
| | - Takashi Kataoka
- Oita University Hospital Rehabilitation Center, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu-city, Oita, 879-5593, Japan
| | - Tatsuya Iwasaki
- Department of Rehabilitation Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu-city, Oita, 879-5593, Japan
| | - Masashi Kataoka
- Physical Therapy Course of Study, Faculty of Welfare and Health Sciences, Oita University, 700 Dannoharu, Oita-city, Oita, 870-1192, Japan.
| | - Hiroshi Tsumura
- Department of Orthopaedic Surgery, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu-city, Oita, 879-5593, Japan
| |
Collapse
|
17
|
Combining sclerostin neutralization with tissue engineering: An improved strategy for craniofacial bone repair. Acta Biomater 2022; 140:178-189. [PMID: 34875361 DOI: 10.1016/j.actbio.2021.11.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023]
Abstract
Scaffolds associated with different types of mesenchymal stromal stem cells (MSC) are extensively studied for the development of novel therapies for large bone defects. Moreover, monoclonal antibodies have been recently introduced for the treatment of cancer-associated bone loss and other skeletal pathologies. In particular, antibodies against sclerostin, a key player in bone remodeling regulation, have demonstrated a real benefit for treating osteoporosis but their contribution to bone tissue-engineering remains uncharted. Here, we show that combining implantation of dense collagen hydrogels hosting wild-type (WT) murine dental pulp stem cells (mDPSC) with weekly systemic injections of a sclerostin antibody (Scl-Ab) leads to increased bone regeneration within critical size calvarial defects performed in WT mice. Furthermore, we show that bone formation is equivalent in calvarial defects in WT mice implanted with Sost knock-out (KO) mDPSC and in Sost KO mice, suggesting that the implantation of sclerostin-deficient MSC similarly promotes new bone formation than complete sclerostin deficiency. Altogether, our data demonstrate that an antibody-based therapy can potentialize tissue-engineering strategies for large craniofacial bone defects and urges the need to conduct research for antibody-enabled local inhibition of sclerostin. STATEMENT OF SIGNIFICANCE: The use of monoclonal antibodies is nowadays broadly spread for the treatment of several conditions including skeletal bone diseases. However, their use to potentialize tissue engineering constructs for bone repair remains unmet. Here, we demonstrate that the neutralization of sclerostin, through either a systemic inhibition by a monoclonal antibody or the implantation of sclerostin-deficient mesenchymal stromal stem cells (MSC) directly within the defect, improves the outcome of a tissue engineering approach, combining dense collagen hydrogels and MSC derived from the dental pulp, for the treatment of large craniofacial bone defects.
Collapse
|
18
|
Kloen P, Loots G, Hamdy R, Smit T. Bridging the gap: compressing non-unions for proper cellular signaling. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
19
|
Mi J, Xu J, Yao Z, Yao H, Li Y, He X, Dai B, Zou L, Tong W, Zhang X, Hu P, Ruan YC, Tang N, Guo X, Zhao J, He J, Qin L. Implantable Electrical Stimulation at Dorsal Root Ganglions Accelerates Osteoporotic Fracture Healing via Calcitonin Gene-Related Peptide. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103005. [PMID: 34708571 PMCID: PMC8728818 DOI: 10.1002/advs.202103005] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/11/2021] [Indexed: 05/18/2023]
Abstract
The neuronal engagement of the peripheral nerve system plays a crucial role in regulating fracture healing, but how to modulate the neuronal activity to enhance fracture healing remains unexploited. Here it is shown that electrical stimulation (ES) directly promotes the biosynthesis and release of calcitonin gene-related peptide (CGRP) by activating Ca2+ /CaMKII/CREB signaling pathway and action potential, respectively. To accelerate rat femoral osteoporotic fracture healing which presents with decline of CGRP, soft electrodes are engineered and they are implanted at L3 and L4 dorsal root ganglions (DRGs). ES delivered at DRGs for the first two weeks after fracture increases CGRP expression in both DRGs and fracture callus. It is also identified that CGRP is indispensable for type-H vessel formation, a biological event coupling angiogenesis and osteogenesis, contributing to ES-enhanced osteoporotic fracture healing. This proof-of-concept study shows for the first time that ES at lumbar DRGs can effectively promote femoral fracture healing, offering an innovative strategy using bioelectronic device to enhance bone regeneration.
Collapse
Affiliation(s)
- Jie Mi
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of OrthopaedicsShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine639 Zhizaoju RoadShanghai200011People's Republic of China
| | - Jian‐Kun Xu
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Zhi Yao
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Hao Yao
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Ye Li
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Xuan He
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Bing‐Yang Dai
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Li Zou
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Wen‐Xue Tong
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Xiao‐Tian Zhang
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Pei‐Jie Hu
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Ye Chun Ruan
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Ning Tang
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Xia Guo
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of OrthopaedicsShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine639 Zhizaoju RoadShanghai200011People's Republic of China
| | - Ju‐Fang He
- Departments of Neuroscience and Biomedical SciencesCity University of Hong KongKowloon Tong999077Hong Kong
| | - Ling Qin
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| |
Collapse
|
20
|
Ali D, Tencerova M, Figeac F, Kassem M, Jafari A. The pathophysiology of osteoporosis in obesity and type 2 diabetes in aging women and men: The mechanisms and roles of increased bone marrow adiposity. Front Endocrinol (Lausanne) 2022; 13:981487. [PMID: 36187112 PMCID: PMC9520254 DOI: 10.3389/fendo.2022.981487] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoporosis is defined as a systemic skeletal disease characterized by decreased bone mass and micro-architectural deterioration leading to increased fracture risk. Osteoporosis incidence increases with age in both post-menopausal women and aging men. Among other important contributing factors to bone fragility observed in osteoporosis, that also affect the elderly population, are metabolic disturbances observed in obesity and Type 2 Diabetes (T2D). These metabolic complications are associated with impaired bone homeostasis and a higher fracture risk. Expansion of the Bone Marrow Adipose Tissue (BMAT), at the expense of decreased bone formation, is thought to be one of the key pathogenic mechanisms underlying osteoporosis and bone fragility in obesity and T2D. Our review provides a summary of mechanisms behind increased Bone Marrow Adiposity (BMA) during aging and highlights the pre-clinical and clinical studies connecting obesity and T2D, to BMA and bone fragility in aging osteoporotic women and men.
Collapse
Affiliation(s)
- Dalia Ali
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, Odense, Denmark
- *Correspondence: Dalia Ali, ; Abbas Jafari,
| | - Michaela Tencerova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Florence Figeac
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Moustapha Kassem
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Dalia Ali, ; Abbas Jafari,
| |
Collapse
|
21
|
Individualized cyclic mechanical loading improves callus properties during the remodelling phase of fracture healing in mice as assessed from time-lapsed in vivo imaging. Sci Rep 2021; 11:23037. [PMID: 34845246 PMCID: PMC8630002 DOI: 10.1038/s41598-021-02368-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 11/12/2021] [Indexed: 01/15/2023] Open
Abstract
Fracture healing is regulated by mechanical loading. Understanding the underlying mechanisms during the different healing phases is required for targeted mechanical intervention therapies. Here, the influence of individualized cyclic mechanical loading on the remodelling phase of fracture healing was assessed in a non-critical-sized mouse femur defect model. After bridging of the defect, a loading group (n = 10) received individualized cyclic mechanical loading (8–16 N, 10 Hz, 5 min, 3 × /week) based on computed strain distribution in the mineralized callus using animal-specific real-time micro-finite element analysis with 2D/3D visualizations and strain histograms. Controls (n = 10) received 0 N treatment at the same post-operative time-points. By registration of consecutive scans, structural and dynamic callus morphometric parameters were followed in three callus sub-volumes and the adjacent cortex showing that the remodelling phase of fracture healing is highly responsive to cyclic mechanical loading with changes in dynamic parameters leading to significantly larger formation of mineralized callus and higher degree of mineralization. Loading-mediated maintenance of callus remodelling was associated with distinct effects on Wnt-signalling-associated molecular targets Sclerostin and RANKL in callus sub-regions and the adjacent cortex (n = 1/group). Given these distinct local protein expression patterns induced by cyclic mechanical loading during callus remodelling, the femur defect loading model with individualized load application seems suitable to further understand the local spatio-temporal mechano-molecular regulation of the different fracture healing phases.
Collapse
|
22
|
Shimatani A, Toyoda H, Orita K, Hirakawa Y, Aoki K, Oh JS, Shirafuji T, Nakamura H. In vivo study on the healing of bone defect treated with non-thermal atmospheric pressure gas discharge plasma. PLoS One 2021; 16:e0255861. [PMID: 34634068 PMCID: PMC8504758 DOI: 10.1371/journal.pone.0255861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/21/2021] [Indexed: 12/21/2022] Open
Abstract
Medical treatment using non-thermal atmospheric pressure plasma (NTAPP) is rapidly gaining recognition. NTAPP is thought to be a new therapeutic method because it could generate highly reactive species in an ambient atmosphere which could be exposed to biological targets (e.g., cells and tissues). If plasma-generated reactive species could stimulate bone regeneration, NTAPP can provide a new treatment opportunity in regenerative medicine. Here, we investigated the impact of NTAPP on bone regeneration using a large bone defect in New Zealand White rabbits and a simple atmospheric pressure plasma (helium microplasma jet). We observed the recovery progress of the large bone defects by X-ray imaging over eight weeks after surgery. The X-ray results showed a clear difference in the occupancy of the new bone of the large bone defect among groups with different plasma treatment times, whereas the new bone occupancy was not substantial in the untreated control group. According to the results of micro-computed tomography analysis at eight weeks, the most successful bone regeneration was achieved using a plasma treatment time of 10 min, wherein the new bone volume was 1.51 times larger than that in the plasma untreated control group. Using H&E and Masson trichrome stains, nucleated cells were uniformly observed, and no inclusion was confirmed, respectively, in the groups of plasma treatment. We concluded the critical large bone defect were filled with new bone. Overall, these results suggest that NTAPP is promising for fracture treatment.
Collapse
Affiliation(s)
- Akiyoshi Shimatani
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hiromitsu Toyoda
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka City University, Osaka, Japan
- * E-mail: (HT); (J-SO)
| | - Kumi Orita
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | | | - Kodai Aoki
- Department of Physical Electronics and Informatics, Graduate School of Engineering, Osaka City University, Osaka, Japan
| | - Jun-Seok Oh
- Department of Physical Electronics and Informatics, Graduate School of Engineering, Osaka City University, Osaka, Japan
- BioMedical Engineering Center, Graduate School of Engineering, Osaka City University, Osaka, Japan
- * E-mail: (HT); (J-SO)
| | - Tatsuru Shirafuji
- Department of Physical Electronics and Informatics, Graduate School of Engineering, Osaka City University, Osaka, Japan
- BioMedical Engineering Center, Graduate School of Engineering, Osaka City University, Osaka, Japan
| | - Hiroaki Nakamura
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
23
|
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
|
24
|
Uemura T, Yano K, Takamatsu K, Miyashima Y, Yasuda H, Konishi S, Nakamura H. Bone healing of distal radius nonunion treated with bridge plating with bone graft substitutes in combination with systemic romosozumab administration: A case report. Jt Dis Relat Surg 2021; 32:526-530. [PMID: 34145834 PMCID: PMC8343859 DOI: 10.52312/jdrs.2021.82661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/20/2021] [Indexed: 11/21/2022] Open
Abstract
Romosozumab is a humanized, anti-sclerostin monoclonal antibody used to treat osteoporosis, which increases bone formation and decreases bone resorption. It enhances fracture healing and systemic romosozumab administration may have therapeutic potentials for accelerating bone healing of even nonunion. Herein, a 61-year-old heavy smoker male with distal radius nonunion who achieved successful bone union by combination therapy of romosozumab and spanning distraction plate fixation with bone graft substitutes was presented. Through the dorsal approach, atrophic comminuted nonunion of the distal radius was sufficiently debrided. Reduction of the distal radius was performed using indirect ligamentotaxis, and a 14-hole locking plate was fixed from the third metacarpal to the radial shaft. A beta (β) tricalcium phosphate block was mainly packed into the substantial metaphyseal bone defect with additional bone graft from the resected ulnar head. Postoperatively, systemic administration of monthly romosozumab was continued for six months. Complete bone union was achieved 20 weeks postoperatively and the plate was, then, removed. Wrist extension and flexion improved to 75o and 55o, respectively, without pain, and grip strength increased 52 weeks postoperatively from 5.5 kg to 22.4 kg. During romosozumab treatment, bone formation marker levels increased rapidly and finally returned to baseline, and bone resorption marker levels remained low. In conclusion, combination of systemic romosozumab administration and grafting β-tricalcium phosphate with bridge plating provides an effective treatment option for difficult cases of comminuted distal radius nonunion with risk factors such as smoking, diabetes, and fragility.
Collapse
Affiliation(s)
- Takuya Uemura
- Department of Orthopaedic Surgery, Osaka General Hospital of West Japan Railway Company, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
Zhu M, Zhang K, Feng L, Lin S, Pan Q, Bian L, Li G. Surface decoration of development-inspired synthetic N-cadherin motif via Ac-BP promotes osseointegration of metal implants. Bioact Mater 2021; 6:1353-1364. [PMID: 33210028 PMCID: PMC7658495 DOI: 10.1016/j.bioactmat.2020.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/25/2020] [Accepted: 11/01/2020] [Indexed: 02/08/2023] Open
Abstract
Research works on the synergistic effect of surface modified bioactive molecules and bone metal implants have been highlighted. N-cadherin is regarded as a key factor in directing cell-cell interactions during the mesenchymal condensation preceding the osteogenesis in the musculoskeletal system. In this study, the N-cadherin mimetic peptide (Cad) was biofunctionalized on the titanium metal surface via the acryloyl bisphosphonate (Ac-BP). To learn the synergistic effect of N-cadherin mimetic peptide, when tethered with titanium substrates, on promoting osteogenic differentiation of the seeded human mesenchymal stem cells (hMSCs) and the osseointegration at the bone-implant interfaces. Results show that the conjugation of N-cadherin mimetic peptide with Ac-BP promoted the osteogenic gene markers expression in the hMSCs. The biofunctionalized biomaterial surfaces promote the expression of the Wnt/β-catenin downstream axis in the attached hMSCs, and then enhance the in-situ bone formation and osseointegration at the bone-implant interfaces. We conclude that this N-cadherin mimetic peptide tethered on Ti surface promote osteogenic differentiation of hMSCs and osseointegration of biomaterial implants in vitro and in vivo. These findings demonstrate the importance of the development-inspired surface bioactivation of metal implants and shed light on the possible cellular mechanisms of the enhanced osseointegration.
Collapse
Affiliation(s)
- Meiling Zhu
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, SAR, Hong Kong, PR China
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, PR China
| | - Kunyu Zhang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, SAR, PR China
| | - Lu Feng
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, SAR, Hong Kong, PR China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, SAR, Hong Kong, PR China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China
| | - Qi Pan
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, SAR, Hong Kong, PR China
| | - Liming Bian
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, SAR, PR China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China
- Centre of Novel Biomaterials, The Chinese University of Hong Kong, Hong Kong, SAR, PR China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, SAR, Hong Kong, PR China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China
| |
Collapse
|
26
|
Liu D, He S, Chen S, Yang L, Yang J, Bao Q, Qin H, Zhao Y, Zong Z. Wnt/β-catenin signalling promotes more effective fracture healing in aged mice than in adult mice by inducing angiogenesis and cell differentiation. Sci Prog 2021; 104:368504211013223. [PMID: 33950750 PMCID: PMC10358591 DOI: 10.1177/00368504211013223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
To investigate whether activating the Wnt/β-catenin signalling pathway differentially promotes fracture healing in aged and adult individuals. CatnbTM2Kem, Catnblox(ex3) and wild-type adult and aged mice were used in this study. The femur was electroporated through a hole with a diameter of 0.6 mm. On the 7th, 14th and 21st days after fracture establishment, repair of the femoral diaphyseal bone was examined using X-ray and CT, the levels of mRNAs related to Wnt/β-catenin signalling were detected using real-time polymerase chain reaction (RT-PCR), and angiogenesis and cell differentiation were observed using immunohistochemistry. The numbers of osteoclasts were determined by TRAP staining. Wnt/β-catenin activation accelerated fracture healing in adult mice, with more pronounced effects on aged mice. Compared with wild-type mice at the corresponding ages, Wnt/β-catenin signalling activation induced higher levels of angiogenesis and cell differentiation in aged mice than in adult mice and promoted fracture healing. The administration of medications targeting Wnt/β-catenin signalling to aged patients may accelerate fracture healing to a greater extent.
Collapse
Affiliation(s)
| | - Sihao He
- Army Medical University, Chongqing, China
| | - Sixu Chen
- Army Medical University, Chongqing, China
| | - Lei Yang
- Army Medical University, Chongqing, China
| | | | | | - Hao Qin
- Army Medical University, Chongqing, China
| | | | | |
Collapse
|
27
|
Zhang L, Jin L, Guo J, Bao K, Hu J, Zhang Y, Hou Z, Zhang L. Chronic Intermittent Hypobaric Hypoxia Enhances Bone Fracture Healing. Front Endocrinol (Lausanne) 2021; 11:582670. [PMID: 33664707 PMCID: PMC7921462 DOI: 10.3389/fendo.2020.582670] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/14/2020] [Indexed: 01/08/2023] Open
Abstract
The effect of chronic intermittent hypobaric hypoxia (CIHH) on bone fracture healing is not elucidated. The present study aimed to investigate the role of CIHH on bone fracture healing and the mechanism. The Sprague-Dawley rats were randomly divided into the CIHH group and control group and monitored for 2, 4, or 8 weeks after femoral fracture surgery. Bone healing efficiency was significantly increased in the CIHH group as evidenced by higher high-density bone volume fractions, higher bone mineral density, higher maximum force, and higher stiffness. Histologically, the CIHH group exhibited superior bone formation, endochondral ossification, and angiogenic ability compared with the control group. The expression of HIF-1α and its downstream signaling proteins VEGF, SDF-1/CXCR4 axis were increased by the CIHH treatment. Moreover, the expression of RUNX2, osterix, and type I collagen in the callus tissues were also up-regulated in the CIHH group. In conclusion, our study demonstrated that CIHH treatment improves fracture healing, increases bone mineral density, and increases bone strength via the activation of HIF-1α and bone production-related genes.
Collapse
Affiliation(s)
- Li Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lin Jin
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jialiang Guo
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Kai Bao
- Department of Orthopaedic Surgery, Hebei Provincial Hospital of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jinglue Hu
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yingze Zhang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Liping Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
28
|
Yamakawa D, Kawase-Koga Y, Fujii Y, Kanno Y, Sato M, Ohba S, Kitaura Y, Kashiwagi M, Chikazu D. Effects of Helioxanthin Derivative-Treated Human Dental Pulp Stem Cells on Fracture Healing. Int J Mol Sci 2020; 21:E9158. [PMID: 33271795 PMCID: PMC7730800 DOI: 10.3390/ijms21239158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 01/05/2023] Open
Abstract
Bone defects affect patients functionally and psychologically and can decrease quality of life. To resolve these problems, a simple and efficient method of bone regeneration is required. Human dental pulp stem cells (DPSCs) have high proliferative ability and multilineage differentiation potential. In our previous study, we reported a highly efficient method to induce osteogenic differentiation using DPSC sheets treated with a helioxanthin derivative (4-(4-methoxyphenyl)pyrido[40,30:4,5]thieno[2,3-b]pyridine-2-carboxamide (TH)) in a mouse calvarial defect model. However, the localization of the DPSCs after transplantation remains unknown. Therefore, in this study, we investigated the localization of transplanted DPSCs in a mouse fracture model. DPSCs were collected from six healthy patients aged 18-29 years, cultured in normal medium (NM), osteogenic medium (OM), or OM with TH, and fabricated them into cell sheets. To evaluate the efficacy of fracture healing using DPSCs treated with OM+TH, and to clarify the localization of the transplanted DPSC sheets in vivo, we transplanted OM+TH-treated DPSC sheets labeled with PKH26 into mouse tibiae fractures. We demonstrated that transplanted OM+TH-treated DPSCs sheets were localized to the fracture site and facilitated bone formation. These results indicated that transplanted OM+TH-treated DPSCs were localized at fracture sites and directly promoted fracture healing.
Collapse
Affiliation(s)
- Daiki Yamakawa
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (D.Y.); (Y.F.); (Y.K.); (M.S.); (D.C.)
| | - Yoko Kawase-Koga
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (D.Y.); (Y.F.); (Y.K.); (M.S.); (D.C.)
- Department of Oral and Maxillofacial Surgery, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Yasuyuki Fujii
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (D.Y.); (Y.F.); (Y.K.); (M.S.); (D.C.)
| | - Yuki Kanno
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (D.Y.); (Y.F.); (Y.K.); (M.S.); (D.C.)
- Department of Oral and Maxillofacial Surgery, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Marika Sato
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (D.Y.); (Y.F.); (Y.K.); (M.S.); (D.C.)
| | - Shinsuke Ohba
- Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan;
| | - Yoshiaki Kitaura
- Department of Bioengineering, School of Engneering, The University of Tokyo, 7-3-1 Hongou, Bunkyo-ku, Tokyo 113-0033, Japan;
| | - Miki Kashiwagi
- Department of Oral-Maxillofacial Surgery and Orthodontics, University of Tokyo Hospital, 7-3-1 Hongou, Bunkyo-ku, Tokyo 113-0033, Japan;
| | - Daichi Chikazu
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (D.Y.); (Y.F.); (Y.K.); (M.S.); (D.C.)
| |
Collapse
|
29
|
Mi J, Xu J, Yao H, Li X, Tong W, Li Y, Dai B, He X, Chow DHK, Li G, Lui KO, Zhao J, Qin L. Calcitonin Gene-Related Peptide Enhances Distraction Osteogenesis by Increasing Angiogenesis. Tissue Eng Part A 2020; 27:87-102. [PMID: 32375579 DOI: 10.1089/ten.tea.2020.0009] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Distraction osteogenesis (DO) is a well-established surgical technique for treating bone defect and limb lengthening. The major drawback of DO is the long treatment period as the external fixator has to be kept in place until consolidation is completed. Calcitonin gene-related peptide (CGRP) has been reported to promote angiogenesis by affecting endothelial progenitor cells (EPCs) in limb ischemia and wound healing. Thus, the goal of this study was to evaluate the angiogenic effect of exogenous CGRP on bone regeneration in a rat DO model. Exogenous CGRP was directly injected into the bone defect after each cycle of distraction in vivo. Microcomputed tomography, biomechanical test, and histological analysis were performed to assess the new bone formation. Angiography and immunofluorescence were performed to assess the formation of blood vessels. CD31+CD144+ EPCs in the bone defect were quantified with flow cytometry. In in vitro study, bone marrow stem cells (BMSCs) were used to investigate the effect of CGRP on EPCs production during endothelial differentiation. Our results showed that CGRP significantly promoted bone regeneration and vessel formation after consolidation. CGRP significantly increased the fraction of CD31+CD144+EPCs and the capillary density in the bone defect at the end of distraction phase. CGRP increased EPC population in the endothelial differentiation of BMSCs in vitro by activating PI3K/AKT signaling pathway. Furthermore, differentiated EPCs rapidly assembled into tube-like structures and promoted osteogenic differentiation of BMSCs. In conclusion, CGRP increased EPC population and promoted blood vessel formation and bone regeneration at the defect region in a DO model. Impact statement Distraction osteogenesis (DO) is a well-established surgical technique for limb lengthening and bone defect. The disadvantage of this technique is that external fixator is needed to be kept in place for about 12 months. This may result in increased risk of infection, financial burden, and negative psychological impacts. In this study, we have injected calcitonin gene-related peptide (CGRP) into the defect region after distraction and found that CGRP enhanced vessel formation and bone regeneration in a rat DO model. This suggests that a controlled delivery system for CGRP could be developed and applied clinically for accelerating bone regeneration in patients with DO.
Collapse
Affiliation(s)
- Jie Mi
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hao Yao
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xisheng Li
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ye Li
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xuan He
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Dick Ho Kiu Chow
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Li
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Kathy O Lui
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
30
|
Comeau-Gauthier M, Tarchala M, Luna JLRG, Harvey E, Merle G. Unleashing β-catenin with a new anti-Alzheimer drug for bone tissue regeneration. Injury 2020; 51:2449-2459. [PMID: 32829895 DOI: 10.1016/j.injury.2020.07.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/18/2020] [Indexed: 02/02/2023]
Abstract
The Wnt/β-catenin signaling pathway is critical for bone differentiation and regeneration. Tideglusib, a selective FDA approved glycogen synthase kinase-3β (GSK-3β) inhibitor, has been shown to promote dentine formation, but its effect on bone has not been examined. Our objective was to study the effect of localized Tideglusib administration on bone repair. Bone healing between Tideglusib treated and control mice was analysed at 7, 14 and 28 days postoperative (PO) with microCT, dynamic histomorphometry and immunohistology. There was a local downregulation of GSK-3β in Tideglusib animals, resulting in a significant increase in the amount of new bone formation with both enhanced cortical bone bridging and medullary bone deposition. The bone formation in the Tideglusib group was characterized by early osteoblast differentiation with down-regulation of GSK-3β at day 7 and 14, and higher accumulation of active β-catenin at day 14. Here, for the first time, we show a positive effect of Tideglusib on bone formation through the inactivation of GSK-3β. Furthermore, the findings suggest that Tideglusib does not interfere with precursor cell recruitment and commitment, contrary to other GSK-3β antagonists such as lithium chloride. Taken together, the results indicate that Tideglusib could be used directly at a fracture site during the initial intraoperative internal fixation without the need for further surgery, injection or drug delivery system. This FDA-approved drug may be useful in the future for the prevention of non-union in patients presenting with a high risk for fracture-healing.
Collapse
Affiliation(s)
- Marianne Comeau-Gauthier
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal, Canada; Experimental Surgery, Faculty of Medicine, McGill University. Rue de la Montaigne, Montreal, QC, Canada.
| | - Magdalena Tarchala
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal, Canada; Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Qc., H3G 1A4 Canada.
| | - Jose Luis Ramirez-Garcia Luna
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal, Canada; Experimental Surgery, Faculty of Medicine, McGill University. Rue de la Montaigne, Montreal, QC, Canada; Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Qc., H3G 1A4 Canada.
| | - Edward Harvey
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal, Canada; Bone Engineering Labs, Montreal General Hospital, 1650 Cedar Avenue, Room C10-124, Montreal, Qc., H3G 1A4 Canada.
| | - Geraldine Merle
- Chemical Engineering Department, Polytechnique J.-A.-Bombardier building Polytechnique Montréal C.P. 6079, succ. Centre-ville, Montréal (Québec), H3C 3A7, Canada.
| |
Collapse
|
31
|
Marmor MT, Dailey H, Marcucio R, Hunt AC. Biomedical research models in the science of fracture healing - Pitfalls & promises. Injury 2020; 51:2118-2128. [PMID: 32591215 DOI: 10.1016/j.injury.2020.06.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/14/2020] [Indexed: 02/02/2023]
Abstract
Development of intervention strategies to stimulate fracture healing has long been a focus of musculoskeletal research. Considerable investment in empirical research has led to the discovery of several genes and signaling pathways that are involved in skeletal development and regeneration. However, there are currently very few biologic interventions that can efficiently be used to enhance fracture healing in clinical practice. This translational barrier is due in part to experimental barriers to mechanism discovery. Animal models, biomechanical models, finite element models, and mathematical models are a few examples of models that aid in the discovery of mechanisms. Understanding the advantages, limitations, and specialized uses of each model type is critical to our ability to interpret mechanistic insights from such research and to help bridge the translation gap between pre-clinical research and clinical practice. In this review, we look at specific modeling methods used in the study of the fracture healing mechanism. We also discuss the strength and limitations to translation of each method, hopefully leading to a better understanding of how we can use models to advance the study of fracture healing.
Collapse
Affiliation(s)
- Meir T Marmor
- Department of Orthopaedic Surgery, Zuckerberg San Francisco General Hospital, Orthopaedic Trauma Institute, University of California, San Francisco, CA, United States.
| | - Hannah Dailey
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA, United States
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, Zuckerberg San Francisco General Hospital, Orthopaedic Trauma Institute, University of California, San Francisco, CA, United States
| | - Anthony C Hunt
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, United States
| |
Collapse
|
32
|
Li X, Xu J, Dai B, Wang X, Guo Q, Qin L. Targeting autophagy in osteoporosis: From pathophysiology to potential therapy. Ageing Res Rev 2020; 62:101098. [PMID: 32535273 DOI: 10.1016/j.arr.2020.101098] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/26/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022]
Abstract
Osteoporosis is a highly prevalent disorder characterized by the loss of bone mass and microarchitecture deterioration of bone tissue, attributed to various factors, including menopause (primary), aging (primary) and adverse effects of relevant medications (secondary). In recent decades, knowledge regarding the etiological mechanisms underpinning osteoporosis emphasizes that bone cellular homeostasis, including the maintenance of cell functions, differentiation, and the response to stress, is tightly regulated by autophagy, which is a cell survival mechanism for eliminating and recycling damaged proteins and organelles. With the important roles in the maintenance of cellular homeostasis and organ function, autophagy has emerged as a potential target for the prevention and treatment of osteoporosis. In this review, we update and discuss the pathophysiology of autophagy in normal bone cell life cycle and metabolism. Then, the alternations of autophagy in primary and secondary osteoporosis, and the accompanied pathological process are discussed. Finally, we discuss current strategies, limitations, and challenges involved in targeting relevant pathways and propose strategies by which such hurdles may be circumvented in the future for their translation into clinical validations and applications for the prevention and treatment of osteoporosis.
Collapse
|
33
|
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
|
34
|
Schemitsch EH, Miclau T, Karachalios T, Nowak LL, Sancheti P, Poolman RW, Caminis J, Daizadeh N, Dent-Acosta RE, Egbuna O, Chines A, Maddox J, Grauer A, Bhandari M. A Randomized, Placebo-Controlled Study of Romosozumab for the Treatment of Hip Fractures. J Bone Joint Surg Am 2020; 102:693-702. [PMID: 31977817 PMCID: PMC7508283 DOI: 10.2106/jbjs.19.00790] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Romosozumab is a bone-forming antibody that increases bone formation and decreases bone resorption. We conducted a double-blinded, randomized, phase-2, dose-finding trial to evaluate the effect of romosozumab on the clinical outcomes of open reduction and internal fixation of intertrochanteric or femoral neck hip fractures. METHODS Patients (55 to 94 years old) were randomized 2:3:3:3 to receive 3 subcutaneous injections of romosozumab (70, 140, or 210 mg) or a placebo postoperatively on day 1 and weeks 2, 6, and 12. The primary end point was the difference in the mean timed "Up & Go" (TUG) score over weeks 6 to 20 for romosozumab versus placebo. Additional end points included the time to radiographic evidence of healing and the score on the Radiographic Union Scale for Hip (RUSH). RESULTS A total of 332 patients were randomized: 243 to receive romosozumab (70 mg, n = 60; 140 mg, n = 93; and 210 mg, n = 90) and 89 to receive a placebo. Although TUG scores improved during the study, they did not differ significantly between the romosozumab and placebo groups over weeks 6 to 20 (p = 0.198). The median time to radiographic evidence of healing was 16.4 to 16.9 weeks across treatment groups. The RUSH scores improved over time across treatment groups but did not differ significantly between the romosozumab and placebo groups. The overall safety and tolerability profile of romosozumab was comparable with that of the placebo. CONCLUSIONS Romosozumab did not improve the fracture-healing-related clinical and radiographic outcomes in the study population. LEVEL OF EVIDENCE Therapeutic Level I. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Emil H. Schemitsch
- Department of Surgery, University of Western Ontario, London, Ontario, Canada,Email address for E.H. Schemitsch:
| | - Theodore Miclau
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California,Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, California
| | - Theofilos Karachalios
- Orthopaedic Department UGHL, School of Health Sciences, University of Thessalia, Larissa, Greece
| | - Lauren L. Nowak
- Department of Surgery, University of Western Ontario, London, Ontario, Canada
| | - Parag Sancheti
- Sancheti Institute of Orthopaedics and Rehabilitation, Pune, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Rothe R, Schulze S, Neuber C, Hauser S, Rammelt S, Pietzsch J. Adjuvant drug-assisted bone healing: Part III - Further strategies for local and systemic modulation. Clin Hemorheol Microcirc 2020; 73:439-488. [PMID: 31177207 DOI: 10.3233/ch-199104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this third in a series of reviews on adjuvant drug-assisted bone healing, further approaches aiming at influencing the healing process are discussed. Local and systemic modulation of bone metabolism is pursued with use of a number of drugs with completely different indications, which are characterized by a pleiotropic spectrum of action. These include drugs used to treat lipid disorders (HMG-CoA reductase inhibitors), hypertension (ACE inhibitors), osteoporosis (bisphosphonates), cancer (proteasome inhibitors) and others. Potential applications to enhance bone healing are discussed.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, Dresden
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
36
|
Çevik HB, Eceviz E, Çilingir Kaya ÖT, Ercan F, Çeçen GS. The effect of topical and systemic tranexamic acid on fracture healing in rats. ACTA ORTHOPAEDICA ET TRAUMATOLOGICA TURCICA 2020; 54:207-212. [PMID: 32254038 DOI: 10.5152/j.aott.2020.02.44] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The aim of the present study was to determine the effect of topical and systemic tranexamic acid (TXA) on fracture healing in a rat surgical model. METHODS We created standard, right-sided, open, diaphyseal femoral fractures with intramedullary Kirschner wire fixation in 48 male rats and divided them into three groups: a topical TXA (10 mg/kg) group, a systemic TXA (10 mg/kg) group, and a control group. Fracture healing was evaluated radiographically and histologically after early (week 2) and late (week 4) postoperative sacrifice. RESULTS The radiological scores differed significantly among the all groups (p=0.001), as did the week 2 and 4 scores (p=0.003 and p=0.010, respectively). Radiologically, the topical TXA group exhibited better bone healing at both 2 (p=0.001) and 4 (p=0.007) weeks than the control group, and the systemic group showed better healing at both 2 (p=0.027) and 4 (p=0.023) weeks than the control TXA group. Moreover, bone healing was better in the group treated with topical rather than systemic TXA on radiological examinations performed at 2 (p=0.001) and 4 (p=0.007) weeks postoperatively (p=0.001 and p=0.007, respectively). Histologically, the groups differed significantly (p=0.001). The histological scores differed significantly among the all groups (p=0.001). At 2 weeks, the topical TXA group exhibited significantly better bone healing than the control group (p=0.001). CONCLUSION Our results suggested that topical application of TXA in fracture patients may accelerate healing, whereas systemic administration may adversely affect healing.
Collapse
Affiliation(s)
- Hüseyin Bilgehan Çevik
- Department of Orthopaedics and Traumatology, University of Health Sciences, Kartal Dr. Lütfi Kırdar Research and Training Hospital, İstanbul, Turkey
| | - Engin Eceviz
- Department of Orthopaedics and Traumatology, University of Health Sciences, Kartal Dr. Lütfi Kırdar Research and Training Hospital, İstanbul, Turkey
| | | | - Feriha Ercan
- Department of Histology and Embryology, Marmara University, School of Medicine, İstanbul, Turkey
| | - Gültekin Sıtkı Çeçen
- Department of Orthopaedics and Traumatology, University of Health Sciences, Kartal Dr. Lütfi Kırdar Research and Training Hospital, İstanbul, Turkey
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Substantial advances have been made in understanding the biological basis of fracture healing. Yet, it is unclear whether the presence of osteoporosis or prior or current osteoporosis therapy influences the healing process or is associated with impaired healing. This review discusses the normal process of fracture healing and the role of osteoporosis and patient-specific factors in relation to fracture repair. RECENT FINDINGS The definitive association of osteoporosis to impaired fracture healing remains inconclusive because of limited evidence addressing this point. eStudies testing anabolic agents in preclinical models of ovariectomized animals with induced fractures have produced mostly positive findings showing enhanced fracture repair. Prospective human clinical trials, although few in number and limited in design and to testing only one anabolic agent, have similarly yielded modestly favorable results. Interest is high for exploring currently available osteoporosis therapies for efficacy in fracture repair. Definitive data supporting their efficacy are essential in achieving approval for this indication.
Collapse
Affiliation(s)
- Cheng Cheng
- Endocrine Research Unit, Department of Medicine, San Francisco Veterans Affairs Medical Center, 1700 Owens Street, Room 369, San Francisco, CA, 94158, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, USA
| | - Dolores Shoback
- Endocrine Research Unit, Department of Medicine, San Francisco Veterans Affairs Medical Center, 1700 Owens Street, Room 369, San Francisco, CA, 94158, USA.
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, USA.
| |
Collapse
|
38
|
Kruck B, Zimmermann EA, Damerow S, Figge C, Julien C, Wulsten D, Thiele T, Martin M, Hamdy R, Reumann MK, Duda GN, Checa S, Willie BM. Sclerostin Neutralizing Antibody Treatment Enhances Bone Formation but Does Not Rescue Mechanically Induced Delayed Healing. J Bone Miner Res 2018; 33:1686-1697. [PMID: 29694687 DOI: 10.1002/jbmr.3454] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/05/2018] [Accepted: 04/16/2018] [Indexed: 12/31/2022]
Abstract
During bone healing, tissue formation processes are governed by mechanical strain. Sost/sclerostin, a key Wnt signaling inhibitor and mechano-sensitive pathway, is downregulated in response to mechanical loading. Sclerostin neutralizing antibody (SclAb) increases bone formation. Nevertheless, it remains unclear whether sclerostin inhibition can rescue bone healing in situations of mechanical instability, which otherwise delay healing. We investigated SclAb's influence on tissue formation in a mouse femoral osteotomy, stabilized with rigid or semirigid external fixation. The different fixations allowed different magnitudes of interfragmentary movement during weight bearing, thereby influencing healing outcome. SclAb or vehicle (veh) was administeredand bone healing was assessed at multiple time points up to day 21 postoperatively by in vivo micro-computed tomography, histomorphometry, biomechanical testing, immunohistochemistry, and gene expression. Our results show that SclAb treatment caused a greater bone volume than veh. However, SclAb could not overcome the characteristic delayed healing of semirigid fixation. Indeed, semirigid fixation resulted in delayed healing with a prolonged endochondral ossification phase characterized by increased cartilage, lower bone volume fraction, and less bony bridging across the osteotomy gap than rigid fixation. In a control setting, SclAb negatively affected later stages of healing under rigid fixation, evidenced by the high degree of endosteal bridging at 21 days in the rigid-SclAb group compared with rigid-veh, indicating delayed fracture callus remodeling and bone marrow reconstitution. Under rigid fixation, Sost and sclerostin expression at the gene and protein level, respectively, were increased in SclAb compared with veh-treated bones, suggesting a negative feedback mechanism. Our results suggest that SclAb could be used to enhance overall bone mass but should be carefully considered in bone healing. SclAb may help to increase bone formation early in the healing process but not during advanced stages of fracture callus remodeling and not to overcome delayed healing in semirigid fixation. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Bettina Kruck
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Elizabeth A Zimmermann
- Research Centre, Shriners Hospital for Children-Canada, Department of Pediatric Surgery, McGill University, Montreal, Canada
| | - Sophie Damerow
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christine Figge
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Catherine Julien
- Research Centre, Shriners Hospital for Children-Canada, Department of Pediatric Surgery, McGill University, Montreal, Canada
| | - Dag Wulsten
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Thiele
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Madge Martin
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Laboratoire Modélisation et Simulation Multi-Echelle, Université Paris-Est Créteil, Paris, France
| | - Reggie Hamdy
- Research Centre, Shriners Hospital for Children-Canada, Department of Pediatric Surgery, McGill University, Montreal, Canada
| | - Marie K Reumann
- Siegfried Weller Institut für Unfallmedizinische Forschung, Berufsgenossenschaftliche Unfallklinik, Eberhard Karls Universität, Tuebingen, Germany
| | - Georg N Duda
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Bettina M Willie
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Research Centre, Shriners Hospital for Children-Canada, Department of Pediatric Surgery, McGill University, Montreal, Canada
| |
Collapse
|
39
|
Makino T, Tsukazaki H, Ukon Y, Tateiwa D, Yoshikawa H, Kaito T. The Biological Enhancement of Spinal Fusion for Spinal Degenerative Disease. Int J Mol Sci 2018; 19:ijms19082430. [PMID: 30126106 PMCID: PMC6121547 DOI: 10.3390/ijms19082430] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/04/2018] [Accepted: 08/14/2018] [Indexed: 12/16/2022] Open
Abstract
In this era of aging societies, the number of elderly individuals who undergo spinal arthrodesis for various degenerative diseases is increasing. Poor bone quality and osteogenic ability in older patients, due to osteoporosis, often interfere with achieving bone fusion after spinal arthrodesis. Enhancement of bone fusion requires shifting bone homeostasis toward increased bone formation and reduced resorption. Several biological enhancement strategies of bone formation have been conducted in animal models of spinal arthrodesis and human clinical trials. Pharmacological agents for osteoporosis have also been shown to be effective in enhancing bone fusion. Cytokines, which activate bone formation, such as bone morphogenetic proteins, have already been clinically used to enhance bone fusion for spinal arthrodesis. Recently, stem cells have attracted considerable attention as a cell source of osteoblasts, promising effects in enhancing bone fusion. Drug delivery systems will also need to be further developed to assure the safe delivery of bone-enhancing agents to the site of spinal arthrodesis. Our aim in this review is to appraise the current state of knowledge and evidence regarding bone enhancement strategies for spinal fusion for degenerative spinal disorders, and to identify future directions for biological bone enhancement strategies, including pharmacological, cell and gene therapy approaches.
Collapse
Affiliation(s)
- Takahiro Makino
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Hiroyuki Tsukazaki
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Yuichiro Ukon
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Daisuke Tateiwa
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Hideki Yoshikawa
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Takashi Kaito
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
40
|
Tian L, Sheng Y, Huang L, Chow DHK, Chau WH, Tang N, Ngai T, Wu C, Lu J, Qin L. An innovative Mg/Ti hybrid fixation system developed for fracture fixation and healing enhancement at load-bearing skeletal site. Biomaterials 2018; 180:173-183. [PMID: 30041069 DOI: 10.1016/j.biomaterials.2018.07.018] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 07/05/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
Magnesium (Mg) is a potential biomaterial suitable for developing biodegradable orthopaedic implants, especially as internal fixators for fracture fixation at non-load bearing skeletal sites. However, Mg alone cannot provide sufficient mechanical support for stable fracture fixation at load bearing sites due to its rapid degradation in the early stage after implantation. In consideration of the strengths and weaknesses of Mg, we developed an innovative magnesium/titanium (Mg/Ti) hybrid fixation system for long bone fracture fixation and investigated the fixation efficacy. The finite element analysis (FEA) results indicated that the Mg/Ti hybrid fixation system provided sufficient mechanical support for fracture fixation at load-bearing skeletal site. As a proof-of-concept, we performed a "Z-shaped" open osteotomy at the mid-shaft of rabbit tibia. For comparison, the animals were divided into two groups: Mg/Ti group (fixated with Mg screws and Ti fixators) and Ti control group (fixated with Ti screws and Ti fixators). The radiographic, four-point bending mechanical test, histological and histomorphometric analysis were postoperatively performed in a temporal manner up to 12 weeks. Both X-ray and micro-CT images of the Mg/Ti group showed a larger callus (14.7% at 3rd week and 24.8% at 6th week, n = 5-7, p < 0.05) in the regions of interest (ROIs) over time, especially at the opposite cortex of the fixation plate. At the 12th week post-operation, the biomechanical test result indicated that the rabbit tibia in the Mg/Ti group healed better and the overall mechanical strength was approximately 3-fold higher (n = 8, p < 0.05) than that at 6th week. Furthermore, the FEA revealed that the Mg/Ti group had a higher mechanical strength (19.5% at week 6 and 31.5% at week 12) at the specified ROI and resulted in an earlier and faster endochondral ossification (68.0% at week 3 and 71.4% at week 6) with a higher expression of osteocalcin (54.0%) and collagen I (34.2%) than the Ti control group (n = 4, p < 0.05). Further evaluation suggested that a higher expression of calcitonin gene-related peptide (CGRP), a known osteogenic neuron peptide, in the fracture callus of the Mg/Ti group might be a major underlying mechanism of enhanced fracture healing attributed to the release of Mg ions during the degradation of Mg screws.
Collapse
Affiliation(s)
- Li Tian
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Yifeng Sheng
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Le Huang
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Dick Ho-Kiu Chow
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Wing Ho Chau
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Ning Tang
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - To Ngai
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Chi Wu
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Jian Lu
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region
| | - Ling Qin
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
41
|
Antibody-Mediated Osseous Regeneration for Bone Tissue Engineering in Canine Segmental Defects. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9508721. [PMID: 29682573 PMCID: PMC5851338 DOI: 10.1155/2018/9508721] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/16/2017] [Accepted: 01/16/2018] [Indexed: 01/29/2023]
Abstract
Among many applications of therapeutic monoclonal antibodies (mAbs), a unique approach for regenerative medicine has entailed antibody-mediated osseous regeneration (AMOR). In an effort to identify a clinically relevant model of craniofacial defect, the present study investigated the efficacy of mAb specific for bone morphogenetic protein- (BMP-) 2 to repair canine segmental mandibular continuity defect model. Accordingly, a 15 mm unilateral segmental defect was created in mandible and fixated with a titanium plate. Anorganic bovine bone mineral with 10% collagen (ABBM-C) was functionalized with 25 μg/mL of either chimeric anti-BMP-2 mAb or isotype-matched mAb (negative control). Recombinant human (rh) BMP-2 served as positive control. Morphometric analyses were performed on computed tomography (CT) and histologic images. Bone densities within healed defect sites at 12 weeks after surgery were 1360.81 ± 10.52 Hounsfield Unit (HU), 1044.27 ± 141.16 HU, and 839.45 ± 179.41 HU, in sites with implanted anti-BMP-2 mAb, rhBMP-2, and isotype mAb groups, respectively. Osteoid bone formation in anti-BMP-2 mAb (42.99% ± 8.67) and rhBMP-2 (48.97% ± 2.96) groups was not significantly different but was higher (p < 0.05) than in sites with isotype control mAb (26.8% ± 5.35). In view of the long-term objective of translational application of AMOR in humans, the results of the present study demonstrated the feasibility of AMOR in a large clinically relevant animal model.
Collapse
|
42
|
Chen S, Zheng L, Zhang J, Wu H, Wang N, Tong W, Xu J, Huang L, Zhang Y, Yang Z, Lin G, Wang X, Qin L. A novel bone targeting delivery system carrying phytomolecule icaritin for prevention of steroid-associated osteonecrosis in rats. Bone 2018; 106:52-60. [PMID: 29030232 DOI: 10.1016/j.bone.2017.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/16/2017] [Accepted: 09/18/2017] [Indexed: 01/06/2023]
Abstract
One of the effective strategies for prevention of steroid-associated osteonecrosis (SAON) is to inhibit bone resorption and fat formation and promote bone formation at osteonecrotic sensitive skeletal sites. We identified a novel phytomolecule that showed positive effects on osteogenesis, anti-bone resorption and anti-adipogenesis in vitro and also developed a bone-targeting delivery system (BTDS) for in vivo experimental study. The study investigated if our innovative synthesized BTDS carrying this phytomolecule would be able to effectively prevent SAON in a rat model. SAON was induced by combined injections of lipopolysaccharide and methylprednisolone. SAON rats were divided into four groups, one SAON untreated control group and three SAON treatment groups with different types of delivery systems (Asp8-liposome-icaritin, liposome-icaritin and Asp8-liposome) for two weeks. SAON lesions were identified and osteoclasts activity, osteogenesis and adipogenesis at these sites were evaluated by immunohistochemistry. Ex vitro study was also designed to evaluate the osteogenic and adipogenic potential of the isolated bone marrow stromal cells (BMSCs) via real-time PCR and histochemical staining. Our results showed that as a bone surface-specific BTDS, Asp8-liposome-icaritin effectively prevented steroids-treated rats from SAON with significantly decreased osteocytes apoptosis, down-regulated osteoclatsogenesis and up-regulated osteogenesis. However, both liposome-icaritin and Asp8-liposome treatment did not show significant efficacy for SAON prevention. In summary, this proof-concept-study showed for the first time that the innovative Asp8-liposome-icaritin BTDS was effective for prevention of SAON in terms of bone resorption prevention, adipogenesis suppression, and bone-formation enhancement.
Collapse
Affiliation(s)
- Shihui Chen
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong, PR China.; Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Jiayong Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, PR China
| | - Heng Wu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong, PR China.; Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, 55455, USA
| | - Nan Wang
- Translational Medicine R&D Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Le Huang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Yifeng Zhang
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, PR China
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, PR China
| | - Ge Lin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Xinluan Wang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong, PR China.; Translational Medicine R&D Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China..
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong, PR China.; Translational Medicine R&D Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China..
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW This review provides a summary of the current knowledge on Sost/sclerostin in cancers targeting the bone, discusses novel observations regarding its potential as a therapeutic approach to treat cancer-induced bone loss, and proposes future research needed to fully understand the potential of therapeutic approaches that modulate sclerostin function. RECENT FINDINGS Accumulating evidence shows that sclerostin expression is dysregulated in a number of cancers that target the bone. Further, new findings demonstrate that pharmacological inhibition of sclerostin in preclinical models of multiple myeloma results in a robust prevention of bone loss and preservation of bone strength, without apparent effects on tumor growth. These data raise the possibility of targeting sclerostin for the treatment of cancer patients with bone metastasis. Sclerostin is emerging as a valuable target to prevent the bone destruction that accompanies the growth of cancer cells in the bone. Further studies will focus on combining anti-sclerostin therapy with tumor-targeted agents to achieve both beneficial skeletal outcomes and inhibition of tumor progression.
Collapse
Affiliation(s)
- Michelle M McDonald
- The Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent's School of Medicine, University of New South Wales, Sydney, Australia
| | - Jesus Delgado-Calle
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
- Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA.
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA.
| |
Collapse
|
44
|
Morse A, McDonald MM, Schindeler A, Peacock L, Mikulec K, Cheng TL, Liu M, Ke HZ, Little DG. Sclerostin Antibody Increases Callus Size and Strength but does not Improve Fracture Union in a Challenged Open Rat Fracture Model. Calcif Tissue Int 2017; 101:217-228. [PMID: 28391431 DOI: 10.1007/s00223-017-0275-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/28/2017] [Indexed: 10/19/2022]
Abstract
Open fractures remain a challenge in orthopedics. Current strategies to intervene are often inadequate, particularly in severe fractures or when treatment is delayed. Sclerostin is a negative regulator of bone growth and sclerostin-neutralizing antibodies (Scl-Ab) can increase bone mass and strength. The application of these antibodies to improve orthopedic repair has shown varied results, and may be dependent on the location and severity of the bony injury. We examined Scl-Ab treatment within an established rat osteotomy model with periosteal stripping analogous to open fracture repair. In one study, Scl-Ab was given 25 mg/kg bi-weekly, either from the time of fracture or from 3 weeks post-fracture up to an end-point of 12 weeks. A second study treated only delayed union open fractures that did not show radiographic union by week 6 post-fracture. Outcome measures included radiographic union, microCT analysis of bone volume and architecture, and histology. In the first study, Scl-Ab given from either 0 or 3 weeks significantly improved callus bone volume (+52%, p < 0.05 and +58%, p < 0.01) at 12 weeks, as well as strength (+48%, p < 0.05 and +70%, p < 0.05). Despite these improvements, union rate was not changed. In the second study treating only established delayed fractures, bony callus volume was similarly increased by Scl-Ab treatment; however, this did not translate to increased biomechanical strength or union improvement. Sclerostin antibody treatment has limited effects on the healing of challenging open fractures with periosteal stripping, but shows the greatest benefits on callus size and strength with earlier intervention.
Collapse
Affiliation(s)
- Alyson Morse
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia
- Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Michelle M McDonald
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia
- Bone Biology Program, The Garvan Institute of Medical Research, Sydney, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia
- Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Lauren Peacock
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia
| | - Kathy Mikulec
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia
| | - Tegan L Cheng
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia
- Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Min Liu
- Amgen Inc, Thousand Oaks, USA
| | | | - David G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.
- Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia.
| |
Collapse
|
45
|
Jacobsen CM. Application of anti-Sclerostin therapy in non-osteoporosis disease models. Bone 2017; 96:18-23. [PMID: 27780792 PMCID: PMC5328800 DOI: 10.1016/j.bone.2016.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/17/2016] [Accepted: 10/20/2016] [Indexed: 12/29/2022]
Abstract
Sclerostin, a known inhibitor of the low density lipoprotein related protein 5 and 6 (LRP5 and LRP6) cell surface signaling receptors, is integral in the maintenance of normal bone mass and strength. Patients with loss of function mutations in SOST or missense mutations in LRP5 that prevent Sclerostin from binding and inhibiting the receptor, have significantly increased bone mass. This observation leads to the development of Sclerostin neutralizing therapies to increase bone mass and strength. Anti-Sclerostin therapy has been shown to be effective at increasing bone density and strength in animal models and patients with osteoporosis. Loss of function of Sost or treatment with a Sclerostin neutralizing antibody improves bone properties in animal models of Osteoporosis Pseudoglioma syndrome (OPPG), likely due to action through the LRP6 receptor, which suggests patients may benefit from these therapies. Sclerostin antibody is effective at improving bone properties in mouse models of Osteogenesis Imperfecta, a genetic disorder of low bone mass and fragility due to type I collagen mutations, in as little as two weeks after initiation of therapy. However, these improvements are due to increases in bone quantity as the quality (brittleness) of bone remains unaffected. Similarly, Sclerostin antibody treatment improves bone density in animal models of other diseases. Sclerostin neutralizing therapies are likely to benefit many patients with genetic disorders of bone, as well as other forms of metabolic bone disease.
Collapse
Affiliation(s)
- Christina M Jacobsen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Boston Children's Hospital, Boston, MA, United States; Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States; Division of Genetics, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
46
|
Kostenuik P, Mirza FM. Fracture healing physiology and the quest for therapies for delayed healing and nonunion. J Orthop Res 2017; 35:213-223. [PMID: 27743449 PMCID: PMC6120140 DOI: 10.1002/jor.23460] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 10/07/2016] [Indexed: 02/04/2023]
Abstract
Delayed healing and nonunion of fractures represent enormous burdens to patients and healthcare systems. There are currently no approved pharmacological agents for the treatment of established nonunions, or for the acceleration of fracture healing, and no pharmacological agents are approved for promoting the healing of closed fractures. Yet several pharmacologic agents have the potential to enhance some aspects of fracture healing. In preclinical studies, various agents working across a broad spectrum of molecular pathways can produce larger, denser and stronger fracture calluses. However, untreated control animals in most of these studies also demonstrate robust structural and biomechanical healing, leaving unclear how these interventions might alter the healing of recalcitrant fractures in humans. This review describes the physiology of fracture healing, with a focus on aspects of natural repair that may be pharmacologically augmented to prevent or treat delayed or nonunion fractures (collectively referred to as DNFs). The agents covered in this review include recombinant BMPs, PTH/PTHrP receptor agonists, activators of Wnt/β-catenin signaling, and recombinant FGF-2. Agents from these therapeutic classes have undergone extensive preclinical testing and progressed to clinical fracture healing trials. Each can promote bone formation, which is important for the stability of bridged calluses, and some but not all can also promote cartilage formation, which may be critical for the initial bridging and subsequent stabilization of fractures. Appropriately timed stimulation of chondrogenesis and osteogenesis in the fracture callus may be a more effective approach for preventing or treating DNFs compared with stimulation of osteogenesis alone. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:213-223, 2017.
Collapse
Affiliation(s)
- Paul Kostenuik
- School of DentistryUniversity of MichiganPhylon Pharma ServicesNewbury ParkCalifornia
| | | |
Collapse
|
47
|
Lau KHW, Rundle CH, Zhou XD, Baylink DJ, Sheng MHC. Conditional deletion of IGF-I in osteocytes unexpectedly accelerates bony union of the fracture gap in mice. Bone 2016; 92:18-28. [PMID: 27519969 DOI: 10.1016/j.bone.2016.08.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 08/03/2016] [Accepted: 08/08/2016] [Indexed: 01/14/2023]
Abstract
This study evaluated the effects of deficient IGF-I expression in osteocytes on fracture healing. Transgenic mice with conditional knockout (cKO) of Igf1 in osteocytes were generated by crossing Dmp1-Cre mice with Igf1 flox mice. Fractures were created on the mid-shaft of tibia of 12-week-old male cKO mice and wild-type (WT) littermates by three-point bending. At 21 and 28days post-fracture healing, the increases in cortical bone mineral density, mineral content, bone area, and thickness, as well as sub-cortical bone mineral content at the fracture site were each greater in cKO calluses than in WT calluses. There were 85% decrease in the cartilage area and >2-fold increase in the number of osteoclasts in cKO calluses at 14days post-fracture, suggesting a more rapid remodeling of endochondral bone. The upregulation of mRNA levels of osteoblast marker genes (cbfa1, alp, Opn, and Ocn) was greater in cKO calluses than in WT calluses. μ-CT analysis suggested an accelerated bony union of the fracture gap in cKO mice. The Sost mRNA level was reduced by 50% and the Bmp2 mRNA level was increased 3-fold in cKO fractures at 14days post-fracture, but the levels of these two mRNAs in WT fractures were unchanged, suggesting that the accelerated fracture repair may in part act through the Wnt and/or BMP signaling. In conclusion, conditional deletion of Igf1 in osteocytes not only did not impair, but unexpectedly enhanced, bony union of the fracture gap. The accelerated bony union was due in part to upregulation of the Wnt and BMP2 signaling in response to deficient osteocyte-derived IGF-I expression, which in turn favors intramembranous over endochondral bone repair.
Collapse
Affiliation(s)
- Kin-Hing W Lau
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA; Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA
| | - Charles H Rundle
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA
| | - Xiao-Dong Zhou
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - David J Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Matilda H-C Sheng
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
48
|
Zhang Y, Xu J, Ruan YC, Yu MK, O'Laughlin M, Wise H, Chen D, Tian L, Shi D, Wang J, Chen S, Feng JQ, Chow DHK, Xie X, Zheng L, Huang L, Huang S, Leung K, Lu N, Zhao L, Li H, Zhao D, Guo X, Chan K, Witte F, Chan HC, Zheng Y, Qin L. Implant-derived magnesium induces local neuronal production of CGRP to improve bone-fracture healing in rats. Nat Med 2016; 22:1160-1169. [PMID: 27571347 PMCID: PMC5293535 DOI: 10.1038/nm.4162] [Citation(s) in RCA: 585] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/13/2016] [Indexed: 12/13/2022]
Abstract
Orthopedic implants containing biodegradable magnesium have been used for fracture repair with considerable efficacy; however, the underlying mechanisms by which these implants improve fracture healing remain elusive. Here we show the formation of abundant new bone at peripheral cortical sites after intramedullary implantation of a pin containing ultrapure magnesium into the intact distal femur in rats. This response was accompanied by substantial increases of neuronal calcitonin gene-related polypeptide-α (CGRP) in both the peripheral cortex of the femur and the ipsilateral dorsal root ganglia (DRG). Surgical removal of the periosteum, capsaicin denervation of sensory nerves or knockdown in vivo of the CGRP-receptor-encoding genes Calcrl or Ramp1 substantially reversed the magnesium-induced osteogenesis that we observed in this model. Overexpression of these genes, however, enhanced magnesium-induced osteogenesis. We further found that an elevation of extracellular magnesium induces magnesium transporter 1 (MAGT1)-dependent and transient receptor potential cation channel, subfamily M, member 7 (TRPM7)-dependent magnesium entry, as well as an increase in intracellular adenosine triphosphate (ATP) and the accumulation of terminal synaptic vesicles in isolated rat DRG neurons. In isolated rat periosteum-derived stem cells, CGRP induces CALCRL- and RAMP1-dependent activation of cAMP-responsive element binding protein 1 (CREB1) and SP7 (also known as osterix), and thus enhances osteogenic differentiation of these stem cells. Furthermore, we have developed an innovative, magnesium-containing intramedullary nail that facilitates femur fracture repair in rats with ovariectomy-induced osteoporosis. Taken together, these findings reveal a previously undefined role of magnesium in promoting CGRP-mediated osteogenic differentiation, which suggests the therapeutic potential of this ion in orthopedics.
Collapse
Affiliation(s)
- Yifeng Zhang
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Ye Chun Ruan
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Mei Kuen Yu
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Micheal O'Laughlin
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Helen Wise
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Di Chen
- Department of Biochemistry, Rush University, Chicago, USA
| | - Li Tian
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Dufang Shi
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Jiali Wang
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Sihui Chen
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Jian Q Feng
- Department of Biomedical Sciences, Baylor College of Dentistry, Texas A&M Health Science Center, Dallas, Texas, USA
| | - Dick Ho Kiu Chow
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Xinhui Xie
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Le Huang
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Shuo Huang
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Kwoksui Leung
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Na Lu
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, PR China
| | - Lan Zhao
- Department of Biochemistry, Rush University, Chicago, USA
| | - Huafang Li
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Dewei Zhao
- Department of Orthopedics, Dalian University Zhongshan Hospital, Dalian, PR China
| | - Xia Guo
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, PR China
| | - Kaiming Chan
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Frank Witte
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hsiao Chang Chan
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing, PR China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
- Translational Medicine Research &Development Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Shenzhen, PR China
| |
Collapse
|
49
|
Kashiwagi M, Hojo H, Kitaura Y, Maeda Y, Aini H, Takato T, Chung UI, Ohba S. Local administration of a hedgehog agonist accelerates fracture healing in a mouse model. Biochem Biophys Res Commun 2016; 479:772-778. [PMID: 27693795 DOI: 10.1016/j.bbrc.2016.09.134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 09/26/2016] [Indexed: 12/29/2022]
Abstract
Bone fracture healing is processed through multiple biological stages including the transition from cartilaginous callus to bony callus formation. Because of its specific, temporal and indispensable functions demonstrated by mouse genetic studies, Hedgehog (Hh) signaling is one of the most potent signaling pathways involved in these processes, but the effect of Hh-signaling activation by small compounds on the repair process had not yet been addressed. Here we examined therapeutic effects of local and one shot-administration of the Hh agonist known as smoothened agonist (SAG) on bone fracture healing in a mouse model. A quantitative analysis with three-dimensional micro-computed tomography showed that SAG administration increased the size of both the cartilaginous callus and bony callus at 14 days after the surgery. A histological analysis showed that SAG administration increased the number of cells expressing a proliferation marker and a chondrocyte marker in cartilaginous callus as well as the cells expressing an osteoblast marker in bony callus. These results indicate that the SAG administration resulted in an enhancement of callus formation during bone fracture healing, which is at least in part mediated by an increase in chondrocyte proliferation in cartilaginous callus and the promotion of bone formation in bony callus. Therapeutic strategies with a SAG-mediated protocol may thus be useful for the treatment of bone fractures.
Collapse
Affiliation(s)
- Miki Kashiwagi
- Department of Sensory and Motor System Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-0033, Japan; Division of Clinical Biotechnology, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hironori Hojo
- Department of Bioengineering, The University of Tokyo Graduate School of Engineering, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yoshiaki Kitaura
- Division of Clinical Biotechnology, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-0033, Japan; Department of Bioengineering, The University of Tokyo Graduate School of Engineering, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yujiro Maeda
- Department of Bioengineering, The University of Tokyo Graduate School of Engineering, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Hailati Aini
- Department of Bioengineering, The University of Tokyo Graduate School of Engineering, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Tsuyoshi Takato
- Department of Sensory and Motor System Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Ung-Il Chung
- Division of Clinical Biotechnology, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-0033, Japan; Department of Bioengineering, The University of Tokyo Graduate School of Engineering, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Shinsuke Ohba
- Division of Clinical Biotechnology, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-0033, Japan; Department of Bioengineering, The University of Tokyo Graduate School of Engineering, Bunkyo-ku, Tokyo, 113-8656, Japan.
| |
Collapse
|
50
|
Lv F, Liu Y, Xu X, Wang J, Ma D, Jiang Y, Wang O, Xia W, Xing X, Yu W, Li M. EFFECTS OF LONG-TERM ALENDRONATE TREATMENT ON A LARGE SAMPLE OF PEDIATRIC PATIENTS WITH OSTEOGENESIS IMPERFECTA. Endocr Pract 2016; 22:1369-1376. [PMID: 27482608 DOI: 10.4158/ep161322.or] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Osteogenesis imperfecta (OI) is a group of inherited diseases characterized by reduced bone mass, recurrent bone fractures, and progressive bone deformities. Here, we evaluate the efficacy and safety of long-term treatment with alendronate in a large sample of Chinese children and adolescents with OI. METHODS In this prospective study, a total of 91 children and adolescents with OI were included. The patients received 3 years' treatment with 70 mg alendronate weekly and 500 mg calcium daily. During the treatment, fracture incidence, bone mineral density (BMD), and serum levels of the bone turnover biomarkers (alkaline phosphatase [ALP] and cross-linked C-telopeptide of type I collagen [β-CTX]) were evaluated. Linear growth speed and parameters of safety were also measured. RESULTS After 3 years of treatment, the mean annual fracture incidence decreased from 1.2 ± 0.8 to 0.2 ± 0.3 (P<.01). BMD at the lumbar spine and femoral neck significantly increased by 74.6% and 39.5%, with their BMD Z-score increasing from -3.0 to 0.1 and from -4.2 to -1.3, respectively (both P<.01 vs. baseline). In addition, serum ALP and β-CTX levels decreased by 35.6% and 44.3%, respectively (both P<.05 vs. baseline). Height significantly increased, but without an obvious increase in its Z-score. Patient tolerance of alendronate was good. CONCLUSION Three years' treatment with alendronate was demonstrated for the first time to significantly reduce fracture incidence, increase lumbar spine and femoral neck BMD, and decrease bone turnover biomarkers in Chinese children and adolescents with OI. ABBREVIATIONS ALP = alkaline phosphatase β-CTX = cross-linked C-telopeptide of type I collagen BMD = bone mineral density BP = bisphosphonate DXA = dual-energy X-ray absorptiometry 25OHD = 25-hydroxyvitamin D OI = osteogenesis imperfecta PTH = parathyroid hormone.
Collapse
|