1
|
Wang YB, Li ZP, Wang P, Wang RB, Ruan YH, Shi Z, Li HY, Sun JK, Mi Y, Li CJ, Zheng PY, Zhang CJ. Iron dysregulation, ferroptosis, and oxidative stress in diabetic osteoporosis: Mechanisms, bone metabolism disruption, and therapeutic strategies. World J Diabetes 2025; 16:106720. [DOI: 10.4239/wjd.v16.i6.106720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/22/2025] [Accepted: 04/16/2025] [Indexed: 06/13/2025] Open
Abstract
Diabetic osteoporosis (DOP) is a common complication in diabetes, driven by hyperglycemia-induced metabolic disturbances, chronic inflammation, and oxidative stress. This review describes the critical role of iron metabolism dysregulation in DOP pathogenesis, focusing on ferroptosis, a novel iron-dependent cell death pathway characterized by lipid peroxidation and reactive oxygen species (ROS) overproduction. Diabetic conditions exacerbate iron overload, impairing osteoblast function and enhancing osteoclast activity, while triggering ferroptosis in bone cells. Ferroptosis not only accelerates osteoblast apoptosis but also amplifies osteoclast-mediated bone resorption, synergistically promoting bone loss. Furthermore, chronic inflammation and oxidative stress disrupt the balance between bone formation and resorption, with elevated pro-inflammatory cytokines (e.g., tumor necrosis factor-α, interleukin-6) and ROS exacerbating cellular dysfunction. Therapeutic strategies targeting iron metabolism (e.g., deferoxamine) and ferroptosis inhibition (e.g., nuclear factor erythroid 2-related factor 2/heme oxygenase-1 pathway activation, antioxidants like melatonin) demonstrate potential to mitigate DOP progression. Future research should prioritize personalized interventions, clinical trials of iron chelators and antioxidants, and mechanistic studies to refine therapeutic approaches. This review provides a comprehensive framework for understanding DOP pathogenesis and highlights innovative strategies to improve bone health in diabetic patients.
Collapse
Affiliation(s)
- Yao-Bin Wang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhi-Peng Li
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Peng Wang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Rui-Bo Wang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yu-Hua Ruan
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhen Shi
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hao-Yu Li
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jin-Ke Sun
- The Third Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yang Mi
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Cheng-Jin Li
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Peng-Yuan Zheng
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Chang-Jiang Zhang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
2
|
Silveira A, Davydok A, Krywka C, Scheel M, Weitkamp T, Fleck C, Shahar R, Zaslansky P. Nanocrystal Compressive Residual Stresses: A Strategy to Strengthen the Bony Spines of Osteocytic and Anosteocytic Fish. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410617. [PMID: 40213975 PMCID: PMC12120745 DOI: 10.1002/advs.202410617] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 03/07/2025] [Indexed: 06/01/2025]
Abstract
Bone is a living tissue in which communicating cells, osteocytes, are assumed to be vital for tissue turnover and adaptation. Interestingly however, most advanced teleost fish do not possess osteocyte-mediated porosity, prompting intriguing questions about alternative material-strategies for these bones to cope with damage. Using advanced imaging techniques, including phase-contrast enhanced (PCE) microtomography (µCT) and nanotomography (nanoCT), X-ray fluorescence (XRF), and diffraction (XRD) tomography, the micro- and nano-architectures of osteocytic zebrafish are compared with anosteocytic medaka fishbone. PCE µCT and Zernike phase-contrast nanoCT showed a lack of porosity in medaka bone and 0.75 - 2.26% osteocytic porosity in zebrafish. Both fish species have similar mineralized collagen fibril arrangements containing calcium (Ca) and traces of strontium (Sr) with increased zinc (Zn) localized on the outer bone regions. Medaka bones exhibit reduced apatite nanocrystal lattice spacings on the outer surfaces. Indeed we find higher compressive residual strains (-0.100 ± 0.02) compared to zebrafish (-0.071 ± 0.03). We propose that medaka bone evolved to replace the mechanosensitive osteocytic network by entrapping protective residual strains between collagen nanofibers and mineral crystals. These strains may enhance fracture toughness while making this nanocomposite well-suited for sustaining repeated loading cycles, thus reducing the metabolic costs associated with housing a large network of cells.
Collapse
Affiliation(s)
- Andreia Silveira
- Department for RestorativePreventive and Pediatric DentistryCharité‐Universitaetsmedizin14197BerlinGermany
| | - Anton Davydok
- Institute of Materials PhysicsHelmholtz‐Zentrum Hereon21502GeesthachtGermany
| | - Christina Krywka
- Institute of Materials PhysicsHelmholtz‐Zentrum Hereon21502GeesthachtGermany
| | | | | | - Claudia Fleck
- Fachgebiet Werkstofftechnik/Chair of Materials Science & EngineeringInstitute of Materials Science and TechnologyFaculty III ‐ Process SciencesTechnische Universität Berlin10623BerlinGermany
| | - Ron Shahar
- Koret School of Veterinary MedicineThe Robert H. Smith Faculty of AgricultureFood and Environmental SciencesHebrew University of JerusalemRehovot76100Israel
| | - Paul Zaslansky
- Department for RestorativePreventive and Pediatric DentistryCharité‐Universitaetsmedizin14197BerlinGermany
| |
Collapse
|
3
|
Chen N, Danalache M, Liang C, Alexander D, Umrath F. Mechanosignaling in Osteoporosis: When Cells Feel the Force. Int J Mol Sci 2025; 26:4007. [PMID: 40362247 PMCID: PMC12071322 DOI: 10.3390/ijms26094007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Bone is a highly mechanosensitive tissue, where mechanical signaling plays a central role in maintaining skeletal homeostasis. Mechanotransduction regulates the balance between bone formation and resorption through coordinated interactions among bone cells. Key mechanosensing structures-including the extracellular/pericellular matrix (ECM/PCM), integrins, ion channels, connexins, and primary cilia, translate mechanical cues into biochemical signals that drive bone adaptation. Disruptions in mechanotransduction are increasingly recognized as an important factor in osteoporosis. Under pathological conditions, impaired mechanical signaling reduces bone formation and accelerates bone resorption, leading to skeletal fragility. Defects in mechanotransduction disrupt key pathways involved in bone metabolism, further exacerbating bone loss. Therefore, targeting mechanotransduction presents a promising pharmacological strategy for osteoporosis treatment. Recent advances have focused on developing drugs that enhance bone mechanosensitivity by modulating key mechanotransduction pathways, including integrins, ion channels, connexins, and Wnt signaling. A deeper understanding of mechanosignaling mechanisms may pave the way for novel therapeutic approaches aimed at restoring bone mass, mechanical integrity, and mechanosensitive bone adaptation.
Collapse
Affiliation(s)
- Nuo Chen
- Department of Orthopedic Surgery, University Hospital Tübingen, 72072 Tübingen, Germany; (N.C.)
| | - Marina Danalache
- Department of Orthopedic Surgery, University Hospital Tübingen, 72072 Tübingen, Germany; (N.C.)
| | - Chen Liang
- Department of Orthopedic Surgery, University Hospital Tübingen, 72072 Tübingen, Germany; (N.C.)
| | - Dorothea Alexander
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Felix Umrath
- Department of Orthopedic Surgery, University Hospital Tübingen, 72072 Tübingen, Germany; (N.C.)
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, 72076 Tübingen, Germany;
| |
Collapse
|
4
|
Faraldi M, Sansoni V, Lombardi G. Recent advances in the role of miRNAs in bone disease. Curr Opin Endocrinol Diabetes Obes 2025:01266029-990000000-00128. [PMID: 40178491 DOI: 10.1097/med.0000000000000914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
PURPOSE OF REVIEW This review explores the potential of circulating miRNAs as diagnostic biomarkers for osteoporosis and bone associated disease, highlighting challenges in translating miRNA findings into clinical practice, including variability in circulating miRNA levels, the need for robust assay methods, and the importance of preanalytical and postanalytical variables. RECENT FINDING Recent finding in miRNA research have identified miRNAs involved in bone cells differentiation, function, and in the regulation of osteoblasts, osteocytes, and osteoclasts. Novel miRNAs associated with osteoporosis, fracture risk, and bone turnover, as well as their utility in distinguishing between primary and secondary forms of osteoporosis have been recently identified. On the other hand, clinical implementation of miRNAs is still limited due to the lack of standardized analytical procedures. SUMMARY miRNAs are noncoding RNA molecules that regulate gene expression, making them key players in complex biological processes, such as bone metabolism. The altered expression of several miRNAs may contribute to bone disorders, including osteoporosis. While significant progress has been made in identifying circulating miRNAs associated with bone disorders, the clinical implementation of miRNA-based diagnostics requires further research and standardization of methods before becoming part of clinical practice.
Collapse
Affiliation(s)
- Martina Faraldi
- Laboratory of Experimental Biochemistry and Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milano, Italy
| | - Veronica Sansoni
- Laboratory of Experimental Biochemistry and Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milano, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milano, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| |
Collapse
|
5
|
Andreev D, Porschitz P. Emerging Roles of Eosinophils in Bone. Curr Osteoporos Rep 2025; 23:17. [PMID: 40183859 PMCID: PMC11971228 DOI: 10.1007/s11914-025-00913-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE OF THE REVIEW Eosinophils are traditionally known for their role in immune defense against parasites and their involvement in various immunopathologies, including eosinophilic airway diseases, eosinophilic dermatoses, and gastrointestinal disorders. However, recent findings from our group and other leading laboratories have broadened this perspective, revealing that eosinophils also play crucial roles in tissue development, homeostasis, and regeneration. This review aims to highlight the regulatory functions of eosinophils within the bone niche and emphasize the importance of further research into their role in bone biology. RECENT FINDINGS Growing evidence suggests that eosinophils are key regulators of bone metabolism, extending beyond their established roles in immunity and inflammation. They contribute to bone homeostasis by inhibiting osteoclast differentiation, helping to prevent excessive bone resorption in osteoporosis and inflammatory arthritis. Additionally, eosinophils may promote osteoblast-mediated bone formation, modulate the mesenchymal and hematopoietic stem cell niche, and contribute to the bone microenvironment by affecting vascularization and extracellular matrix composition. However, their impact may vary under pathological conditions. Patients with eosinophilic disorders are often at an increased risk of osteoporosis and fragility fractures, though this is largely attributed to disease-related treatments rather than eosinophil activity itself. Despite emerging insights into the role of eosinophils in bone biology, the underlying mechanisms remain incompletely understood. Further research is essential to elucidate how eosinophils influence bone physiology and pathology.
Collapse
Affiliation(s)
- Darja Andreev
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, 01307, Dresden, Germany.
| | - Pauline Porschitz
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, 01307, Dresden, Germany
| |
Collapse
|
6
|
Tilton M, Liao J, Kim C, Shaygani H, Potes MA, Cordova DJ, Kirkland JL, Miller KM. Tracing Cellular Senescence in Bone: Time-Dependent Changes in Osteocyte Cytoskeleton Mechanics and Morphology. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408517. [PMID: 40026102 PMCID: PMC11985287 DOI: 10.1002/smll.202408517] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/22/2025] [Indexed: 03/04/2025]
Abstract
Aging-related bone loss significantly impacts the growing elderly population globally, leading to debilitating conditions such as osteoporosis. Senescent osteocytes play a crucial role in the aging process of bone. This longitudinal study examines the impact of continuous local and paracrine exposure to senescence-associated secretory phenotype (SASP) factors on biophysical and biomolecular markers in osteocytes. Significant cytoskeletal stiffening in irradiated (IR) osteocytes are found, accompanied by expansion of F-actin areas and a decline in dendritic integrity. These changes, correlating with alterations in pro-inflammatory cytokine levels and osteocyte-specific gene expression, support the reliability of biophysical markers for identifying senescent osteocytes. Notably, local accumulation of SASP factors have a more pronounced impact on osteocyte biophysical properties than paracrine effects, suggesting that the interplay between local and paracrine exposure can substantially influence cellular aging. This study underscores the importance of osteocyte mechanical and morphological properties as biophysical markers of senescence, highlighting their time dependence and differential effects of local and paracrine SASP exposure. Collectively, the investigation into biophysical senescence markers offers unique and reliable functional hallmarks for the non-invasive identification of senescent osteocytes, providing insights that can inform therapeutic strategies to mitigate aging-related bone loss.
Collapse
Affiliation(s)
- Maryam Tilton
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Junhan Liao
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Chanul Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Hossein Shaygani
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Maria Astudillo Potes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Domenic J. Cordova
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - James L. Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kyle M. Miller
- Department of Radiation Oncology Emory University School of Medicine Atlanta, GA 30307, USA
| |
Collapse
|
7
|
Estévez M, Batoni E, Cicuéndez M, Bonatti AF, Fernández-Marcelo T, De Maria C, González B, Izquierdo-Barba I, Vozzi G. Fabrication of 3D Biofunctional Magnetic Scaffolds by Combining Fused Deposition Modelling and Inkjet Printing of Superparamagnetic Iron Oxide Nanoparticles. Tissue Eng Regen Med 2025:10.1007/s13770-025-00711-2. [PMID: 40100619 DOI: 10.1007/s13770-025-00711-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Recently, magnetic composite biomaterials have raised attention in bone tissue engineering as the application of dynamic magnetic fields proved to modulate the proliferation and differentiation of several cell types. METHODS This study presents a novel method to fabricate biofunctional magnetic scaffolds by the deposition of superparamagnetic iron oxide nanoparticles (SPIONs) through thermal Drop-On-Demand inkjet printing on three-dimensional (3D) printed scaffolds. Firstly, 3D scaffolds based on thermoplastic polymeric composed by poly-L-lactic acid/poly-caprolactone/poly(3-hydroxybutyrate-co-3-hydroxyvalerate) were fabricated by Fused Deposition Modelling. Then, in a second step, SPIONs were incorporated onto the surface of the scaffolds by inkjet printing following a designed 2D pattern. RESULTS A complete characterization of the resulting magnetic scaffolds was carried out attending to the surface SPIONs deposits, demonstrating the accuracy and versatility of the production technique, as well as the stability under physiological conditions and the magnetic properties. Biological evaluation with human bone marrow mesenchymal stems cells demonstrated biocompatibility of the scaffolds and increased osteogenic capability under the application of a magnetic field, due to the activation of mechanotransduction processes. CONCLUSION These results show that the developed 3D magnetic biofunctional scaffolds can be a very promising tool for advanced and personalised bone regeneration treatments.
Collapse
Affiliation(s)
- Manuel Estévez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Instituto de Investigación Sanitaria, Universidad Complutense de Madrid, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
| | - Elisa Batoni
- Department of Information Engineering, University of Pisa, Via Girolamo Caruso 16, 56122, Pisa, Italy
- Research Center "E. Piaggio", University of Pisa, Via Largo Lucio Lazzarino 1, 56122, Pisa, Italy
| | - Mónica Cicuéndez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Amedeo Franco Bonatti
- Research Center "E. Piaggio", University of Pisa, Via Largo Lucio Lazzarino 1, 56122, Pisa, Italy
| | - Tamara Fernández-Marcelo
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040, Madrid, Spain
| | - Carmelo De Maria
- Department of Information Engineering, University of Pisa, Via Girolamo Caruso 16, 56122, Pisa, Italy.
- Research Center "E. Piaggio", University of Pisa, Via Largo Lucio Lazzarino 1, 56122, Pisa, Italy.
| | - Blanca González
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Instituto de Investigación Sanitaria, Universidad Complutense de Madrid, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029, Madrid, Spain
| | - Isabel Izquierdo-Barba
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Instituto de Investigación Sanitaria, Universidad Complutense de Madrid, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029, Madrid, Spain.
| | - Giovanni Vozzi
- Department of Information Engineering, University of Pisa, Via Girolamo Caruso 16, 56122, Pisa, Italy
- Research Center "E. Piaggio", University of Pisa, Via Largo Lucio Lazzarino 1, 56122, Pisa, Italy
| |
Collapse
|
8
|
Wang R, Mehrjou B, Dehghan‐Banian D, Wang BYH, Li Q, Deng S, Liu C, Zhang Z, Zhu Y, Wang H, Li D, Lu X, Cheng JCY, Ong MTY, Chan HF, Li G, Chu PK, Lee WYW. Targeting Long Noncoding RNA H19 in Subchondral Bone Osteocytes and the Alleviation of Cartilage Degradation in Osteoarthritis. Arthritis Rheumatol 2025; 77:283-297. [PMID: 39482250 PMCID: PMC11865692 DOI: 10.1002/art.43028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVE Emerging evidence suggests long noncoding RNA H19 is associated with osteoarthritis (OA) pathology. However, how H19 contributes to OA has not been reported. This study aims to investigate the biologic function of H19 in OA subchondral bone remodeling and OA progression. METHODS Clinical joint samples and OA animal models induced by surgical destabilization of the medial meniscus (DMM) were used to verify the causal relationship between osteocyte H19 and OA subchondral bone and cartilage changes. MLO-Y4 osteocyte cells subjected to fluid shear stress were used to verify the mechanism underlying H19-mediated mechanoresponse. Finally, the antisense oligonucleotide (ASO) against H19 was delivered to mice knee joints by magnetic metal-organic framework (MMOF) nanoparticles to develop a site-specific delivery method for targeting osteocyte H19 for OA treatment. RESULTS Both clinical OA subchondral bone and wildtype mice with DMM-induced OA exhibit aberrant higher subchondral bone mass, with more H19 mice expressing osteocytes. On the contrary, mice with osteocyte-specific deletion of H19 are less vulnerable to DMM-induced OA phenotype. In MLO-Y4 cells, H19-mediated osteocyte mechanoresponse through PI3K/AKT/GSK3 signal activation by EZH2-induced H3K27me3 regulation on protein phosphatase 2A inhibition. Targeted inhibition of H19 (using ASO-loaded MMOF) substantially alleviates subchondral bone remodeling and OA phenotype. CONCLUSION In summary, our results provide new evidence that the elevated H19 expression in osteocytes may contribute to aberrant subchondral bone remodeling and OA progression. H19 appears to be required for the osteocyte response to mechanical stimulation, and targeting H19 represents a new promising approach for OA treatment.
Collapse
Grants
- 2020 Rising Star Award, American Society for Bone and Mineral Research
- AoE/M-402/20 Area of Excellence, University Grants Committee, Hong Kong SAR
- Start-up Fund, The Chinese University of Hong Kong, Hong Kong SAR
- Matching Grant Scheme, University Grants Committee, Hong Kong SAR
- 2412162 General Research Fund, University Grants Committee, Hong Kong SAR
- CT1.1 Center for Neuromusculoskeletal Restorative Medicine, Health@InnoHK Program, Innovation Technology Commission, Hong Kong SAR
Collapse
Affiliation(s)
- Rongliang Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Babak Mehrjou
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong Kong SARChina
| | - Dorsa Dehghan‐Banian
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Belle Yu Hsuan Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and Center for Neuromusculoskeletal Restorative MedicineCUHK InnoHK Centres, Hong Kong Science ParkHong Kong SARChina
| | - Qiangqiang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Shuai Deng
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Chuanhai Liu
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Zhe Zhang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and SH Ho Scoliosis Research LaboratoryJoint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, ShatinHong Kong SARChina
| | - Yanlun Zhu
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Haixing Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Dan Li
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong Kong SARChina
| | - Xiaomin Lu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Jack Chun Yiu Cheng
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and SH Ho Scoliosis Research LaboratoryJoint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, ShatinHong Kong SARChina
| | - Michael Tim Yun Ong
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales HospitalThe Chinese University of Hong Kong, ShatinHong Kong SARChina
| | - Hon Fai Chan
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Gang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China, and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Paul K. Chu
- Department of Physics, Department of Materials Science and Engineering, Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong Kong SARChina
| | - Wayne Yuk Wai Lee
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; and Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science ParkHong Kong SARChina
| |
Collapse
|
9
|
Zhang J, Acosta FM, Wang X, Zhao D, Zhang L, Hua R, Guo Q, Zhong L, Qin L, Riquelme MA, Jiang JX. Osteocyte connexin hemichannels and prostaglandin E 2 release dictate bone marrow mesenchymal stromal cell commitment. Proc Natl Acad Sci U S A 2025; 122:e2412144122. [PMID: 39937859 PMCID: PMC11848350 DOI: 10.1073/pnas.2412144122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025] Open
Abstract
Bone is a dynamic organ constantly undergoing remodeling with both bone formation and resorption. Bone formation is mediated by osteoblasts originating from the differentiation of bone marrow (BM) mesenchymal stem and progenitor cells (BM-MSPCs). However, how bone cells communicate with BM-MSPCs to coordinate bone formation remains largely elusive. Here, we unveil a key role of osteocyte connexin 43 (Cx43) hemichannels in regulating the lineage commitment of BM-MSPCs. Two transgenic mouse models expressing dominant negative Cx43 mutants in osteocytes were used: R76W (inhibiting gap junctions) and Δ130 to 136 (inhibiting both hemichannels and gap junctions). BM-MSPCs from Δ130 to 136 mice showed enhanced adipogenic differentiation and reduced osteogenic potential, leading to increased BM adipocytes. Flow cytometry and single-cell RNA sequencing revealed shifts in BM-MSPC subsets, less osteogenic-biased MSPCs, and more adipogenic-biased MSPCs in Δ130 to 136 mice. Conversely, R76W, with more functional hemichannels, exhibited effects similar to WT mice or even greater opposite effects than Δ130 to 136 mice. Prostaglandin E2 (PGE2), released from active Cx43 hemichannels, inhibited adipogenesis and promoted osteogenesis via the PGE2 receptor EP4 and ERK1/2 signaling. Inhibition of Cx43 hemichannels or EP4 led to increased adipogenic-biased MSPCs. Moreover, administration of a Cx43(M1) antibody, which inhibits hemichannels, substantially increased BM adipocytes, accompanied by increased adipogenic-biased MSPCs, and decreased osteogenic-biased MSPCs. Our study highlights the pivotal role of osteocyte Cx43 hemichannels in BM-MSPC fate decision through PGE2 release, providing insights into the precise and highly regulated communication between matrix-bound bone cells and BM-MSPCs, which dictates bone formation and remodeling.
Collapse
Affiliation(s)
- Jingruo Zhang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX78229
| | - Francisca M. Acosta
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX78229
| | - Xuewei Wang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX78229
| | - Dezhi Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX78229
| | - Lidan Zhang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX78229
| | - Rui Hua
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX78229
| | - Qianjin Guo
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX78229
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Manuel A. Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX78229
| | - Jean X. Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX78229
| |
Collapse
|
10
|
Hong GAT, Tobalske BW, van Staaveren N, Leishman EM, Widowski TM, Powers DR, Harlander A. Hen-durance training-effects of an exercise regimen on laying hen muscle architecture and fracture prevalence. ROYAL SOCIETY OPEN SCIENCE 2025; 12:241191. [PMID: 40012755 PMCID: PMC11858752 DOI: 10.1098/rsos.241191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/24/2024] [Accepted: 12/11/2024] [Indexed: 02/28/2025]
Abstract
Domestic chickens kept for egg laying navigate inclines such as ramps in some commercial housing systems to aid in transitions between housing tiers. Laying hens use their wings and hindlimbs in a locomotion called wing-assisted incline running (WAIR) to ascend steep inclines. There is a potential relationship between the strength of the main flight muscles and the health of the keel bone from which they originate. We sought to test the effects of a controlled, WAIR-based exercise regimen during rearing on keel bone health and muscle properties of white- and brown-feathered laying hens. The WAIR exercise regimen, which consisted of exercise twice weekly for 16 weeks did not promote increases in muscle mass or physiological cross-sectional area at 21 weeks of age (WOA) and did not provide long-term benefits on keel fracture prevalence at 40 WOA. However, the brown-feathered birds were found to have lower amounts of keel fractures at 40 WOA in comparison with the white-feathered birds. Future studies should test for training that begins before chicks become fully feathered, exercises that emphasize full excursion of the wing during downstroke and different levels of intensity, frequency and duration to optimize flight muscle architecture and promote keel bone health.
Collapse
Affiliation(s)
- Grace A. T. Hong
- Department of Animal Biosciences, Campbell Centre for the Study of Animal Welfare, University of Guelph, Guelph, OntarioN1G 2W1, Canada
- Department of Animal Biosciences, Centre for Nutrition Modelling, University of Guelph, Guelph, OntarioN1G 2W1, Canada
| | - Bret W. Tobalske
- Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT59812, USA
| | - Nienke van Staaveren
- Department of Animal Biosciences, Campbell Centre for the Study of Animal Welfare, University of Guelph, Guelph, OntarioN1G 2W1, Canada
- Department Population Health Sciences, Utrecht University, Heidelberglaan 8, Utrecht3584 CS, The Netherlands
| | - Emily M. Leishman
- Department of Animal Biosciences, Campbell Centre for the Study of Animal Welfare, University of Guelph, Guelph, OntarioN1G 2W1, Canada
- Department of Animal Biosciences, Centre for Nutrition Modelling, University of Guelph, Guelph, OntarioN1G 2W1, Canada
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, OntarioN1G 2W1, Canada
| | - Tina M. Widowski
- Department of Animal Biosciences, Campbell Centre for the Study of Animal Welfare, University of Guelph, Guelph, OntarioN1G 2W1, Canada
| | - Donald R. Powers
- Department of Biology, George Fox University, 414N Meridian Street, Newberg, OR97132, USA
| | - Alexandra Harlander
- Department of Animal Biosciences, Campbell Centre for the Study of Animal Welfare, University of Guelph, Guelph, OntarioN1G 2W1, Canada
| |
Collapse
|
11
|
Rabbitt D, Villapún VM, Carter LN, Man K, Lowther M, O'Kelly P, Knowles AJ, Mottura A, Tang YT, Luerti L, Reed RC, Cox SC. Rethinking Biomedical Titanium Alloy Design: A Review of Challenges from Biological and Manufacturing Perspectives. Adv Healthc Mater 2025; 14:e2403129. [PMID: 39711273 PMCID: PMC11804846 DOI: 10.1002/adhm.202403129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/14/2024] [Indexed: 12/24/2024]
Abstract
Current biomedical titanium alloys have been repurposed from other industries, which has contributed to several biologically driven implant failure mechanisms. This review highlights the added value that may be gained by building an appreciation of implant biological responses at the onset of alloy design. Specifically, the fundamental mechanisms associated with immune response, angiogenesis, osseointegration and the potential threat of infection are discussed, including how elemental selection can modulate these pivotal systems. With a view to expedite inclusion of these interactions in alloy design criteria, methods to analyze these performance characteristics are also summarized. While machine learning techniques are being increasingly used to unearth complex relationships between alloying elements and material properties, much is still unknown about the correlation between composition and some bio-related properties. To bridge this gap, high-throughput methods are also reviewed to validate biological response along with cutting edge manufacturing approaches that may support rapid discovery. Taken together, this review encourages the alloy development community to rethink their approach to enable a new generation of biomedical implants intrinsically designed for a life in the body, including functionality to tackle biological challenges thereby offering improved patient outcomes.
Collapse
Affiliation(s)
- Daisy Rabbitt
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | - Victor M. Villapún
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | - Luke N. Carter
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | - Kenny Man
- Department of Oral and Maxillofacial Surgery & Special Dental CareUniversity Medical Center UtrechtUtrecht3508 GAThe Netherlands
- Regenerative Medicine Center UtrechtUniversity Medical Center UtrechtUtrecht3584 CTThe Netherlands
| | - Morgan Lowther
- Paihau‐Robinson Research InstituteVictoria University of WellingtonWellington5010New Zealand
| | - Paraic O'Kelly
- Center for the Accelerated Maturation of MaterialsDepartment of Materials Science and EngineeringThe Ohio State University1305 Kinnear RoadColumbusOH43212USA
| | | | - Alessandro Mottura
- School of Metallurgy and MaterialsUniversity of BirminghamBirminghamB15 2TTUK
| | - Yuanbo T. Tang
- School of Metallurgy and MaterialsUniversity of BirminghamBirminghamB15 2TTUK
| | - Lorenzo Luerti
- Alloyed LtdUnit 15, Oxford Industrial ParkYarntonOX5 1QUUK
| | - Roger C. Reed
- School of Metallurgy and MaterialsUniversity of BirminghamBirminghamB15 2TTUK
- Department of MaterialsUniversity of OxfordParks RoadOxfordOX1 3PJUK
| | - Sophie C. Cox
- School of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| |
Collapse
|
12
|
Kanai T, Osawa K, Kajiwara K, Sato Y, Sawa Y. Study of Podoplanin-Deficient Mouse Bone with Mechanical Stress. Dent J (Basel) 2025; 13:61. [PMID: 39996935 PMCID: PMC11854086 DOI: 10.3390/dj13020061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 02/26/2025] Open
Abstract
Objective: We investigated morphological differences in osteocyte processes between aged mice and our original podoplanin-conditional knockout (cKO) mice in which the floxed exon 3 of podoplanin was deleted by Dmp-1-driven Cre (Dmp1-Cre;PdpnΔ/Δ). Methods: SEM observation on osteocyte cell process, histochemistry for bone remodeling with mechanostress, and RT-PCR for RANKL and M-CSF in podoplanin cKO mouse bone with mechanostress was investigated. Results: SEM observations showed fewer and thinner osteocyte processes in femurs from 23-week-old Dmp1-Cre;PdpnΔ/Δ mice than from 23-week-old wild-type mice, while the numbers of osteocyte processes in femurs and calvarias were similar in 23-week-old Dmp1-Cre;PdpnΔ/Δ mice and 48-week-old wild-type mice. Furthermore, cell process numbers in femurs and calvarias were significantly smaller in 23-week-old Dmp1-Cre;PdpnΔ/Δ mice than in 48-week-old wild-type mice. In the test for differences in alveolar bone resorption under mechanical stress between Dmp1-Cre;PdpnΔ/Δ and wild-type mice, the area of TRAP-positive resorption pits was larger in wild-type mice than in Dmp1-Cre;PdpnΔ/Δ mice. In a quantitative tissue PCR analysis, the mRNA expression levels of RANKL and M-CSF in alveolar bone under mechanical stress were significantly lower in Dmp1-Cre;PdpnΔ/Δ mice than in wild-type mice. These results suggest that a reduction in cell process formation in osteocytes with podoplanin cKO affected the absorption of alveolar bone under mechanical stress in Dmp1-Cre;PdpnΔ/Δ mice. Conclusions: In podoplanin-deficient bone, the deformation of osteocyte processes by mechanical stimuli is not recognized as a stress due to the lower number of cell processes with podoplanin deficiency; therefore, the production of osteoclast migration/differentiation factors by activated osteocytes is not fully induced and macrophage migration to alveolar bone with mechanical stress appeared to be suppressed. These results indicate that podoplanin-dependent osteocyte process formation indirectly plays a key role in sensing mechanical stress in bone.
Collapse
Affiliation(s)
- Takenori Kanai
- Department of Orthodontics, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13 Nishi 7, Kita-ku, Sapporo 060-8586, Japan; (T.K.); (K.O.); (Y.S.)
| | - Kyoko Osawa
- Department of Orthodontics, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13 Nishi 7, Kita-ku, Sapporo 060-8586, Japan; (T.K.); (K.O.); (Y.S.)
| | - Koichiro Kajiwara
- Department of Oral Growth & Development, Hokkaido University, Kita 13 Nishi 7, Kita-ku, Sapporo 060-8586, Japan;
| | - Yoshiaki Sato
- Department of Orthodontics, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita 13 Nishi 7, Kita-ku, Sapporo 060-8586, Japan; (T.K.); (K.O.); (Y.S.)
| | - Yoshihiko Sawa
- Department of Oral Function & Anatomy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-0914, Japan
| |
Collapse
|
13
|
Narain R, Muncie-Vasic JM, Weaver VM. Forcing the code: tension modulates signaling to drive morphogenesis and malignancy. Genes Dev 2025; 39:163-181. [PMID: 39638568 PMCID: PMC11789492 DOI: 10.1101/gad.352110.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Development and disease are regulated by the interplay between genetics and the signaling pathways stimulated by morphogens, growth factors, and cytokines. Experimental data highlight the importance of mechanical force in regulating embryonic development, tissue morphogenesis, and malignancy. Force not only sculpts tissue movements to drive embryogenesis and morphogenesis but also modifies the context of biochemical signaling and gene expression to regulate cell and tissue fate. Not surprisingly, experiments have demonstrated that perturbations in cell tension drive malignancy and metastasis by altering biochemical signaling and gene expression through modifications in cytoskeletal tension, transmembrane receptor structure and function, and organelle phenotype that enhance cell growth and survival, alter metabolism, and foster cell migration and invasion. At the tissue level, tumor-associated forces disrupt cell-cell adhesions to perturb tissue organization, compromise vascular integrity to induce hypoxia, and interfere with antitumor immunity to foster metastasis and treatment resistance. Exciting new approaches now exist with which to clarify the relationship between mechanotransduction, biochemical signaling, and gene expression in development and disease. Indeed, gaining insight into these interactions is essential to unravel molecular mechanisms that regulate development and clarify the molecular basis of cancer.
Collapse
Affiliation(s)
- Radhika Narain
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California 94720, USA
| | | | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA;
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94143, USA
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, California 94143
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
14
|
Shariati K, Bedar M, Huang KX, Moghadam S, Mirzaie S, LaGuardia JS, Chen W, Kang Y, Ren X, Lee JC. Biomaterial Cues for Regulation of Osteoclast Differentiation and Function in Bone Regeneration. ADVANCED THERAPEUTICS 2025; 8:2400296. [PMID: 39867107 PMCID: PMC11756815 DOI: 10.1002/adtp.202400296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Indexed: 01/28/2025]
Abstract
Tissue regeneration involves dynamic dialogue between and among different cells and their surrounding matrices. Bone regeneration is specifically governed by reciprocity between osteoblasts and osteoclasts within the bone microenvironment. Osteoclast-directed resorption and osteoblast-directed formation of bone are essential to bone remodeling, and the crosstalk between these cells is vital to curating a sequence of events that culminate in the creation of bone tissue. Among bone biomaterial strategies, many have investigated the use of different material cues to direct the development and activity of osteoblasts. However, less attention has been given to exploring features that similarly target osteoclast formation and activity, with even fewer strategies demonstrating or integrating biomaterial-directed modulation of osteoblast-osteoclast coupling. This review aims to describe various biomaterial cues demonstrated to influence osteoclastogenesis and osteoclast function, emphasizing those that enhance a material construct's ability to achieve bone healing and regeneration. Additionally discussed are approaches that influence the communication between osteoclasts and osteoblasts, particularly in a manner that takes advantage of their coupling. Deepening our understanding of how biomaterial cues may dictate osteoclast differentiation, function, and influence on the microenvironment may enable the realization of bone-replacement interventions with enhanced integrative and regenerative capacities.
Collapse
Affiliation(s)
- Kaavian Shariati
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Meiwand Bedar
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Kelly X. Huang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Shahrzad Moghadam
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Sarah Mirzaie
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Jonnby S. LaGuardia
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Wei Chen
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Youngnam Kang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Xiaoyan Ren
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Justine C. Lee
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
- Department of Orthopaedic Surgery, Los Angeles, CA, 90095, USA
- UCLA Molecular Biology Institute, Los Angeles, CA, 90095, USA
| |
Collapse
|
15
|
Yang XD, Haga CL, Phinney DG. Signaling Dynamics in Osteogenesis: Unraveling Therapeutic Targets for Bone Generation. Curr Drug Targets 2025; 26:350-366. [PMID: 39791147 DOI: 10.2174/0113894501359782241216082049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025]
Abstract
Diseases affecting bone encompass a spectrum of disorders, from prevalent conditions such as osteoporosis and Paget's disease, collectively impacting millions, to rare genetic disorders including Fibrodysplasia Ossificans Progressiva (FOP). While several classes of drugs, such as bisphosphonates, synthetic hormones, and antibodies, are utilized in the treatment of bone diseases, their efficacy is often curtailed by issues of tolerability and high incidence of adverse effects. Developing therapeutic agents for bone diseases is hampered by the fact that numerous pathways regulating bone metabolism also perform pivotal functions in other organ systems. Consequently, the selection of an appropriate target is a complicated process despite the significant demand for novel medications to address bone diseases. Research has shown the role of various cell signaling pathways, including Wnt, PTHR1, CASR, BMPRs, OSCAR, and TWIST1, in the regulation of osteogenesis, bone remodeling, and homeostasis. Disruptions in bone homeostasis can result in decreased bone density and the onset of osteoporosis. There remains a need for the development of drugs that can enhance bone remodeling with improved side effects profiles. The exploration of promising targets to stimulate bone formation has the potential to significantly advance the field of bone-related medical care, thereby improving the quality of life for millions. Additionally, a deeper understanding of anabolic and catabolic pathway mechanisms could enable future studies to explore synergistic effects between unrelated pathways. Herein, we explore potential drug targets that may be exploited therapeutically using small molecule agonists or antagonists to promote bone remodeling and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Xue D Yang
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL33458, USA
| | - Christopher L Haga
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL33458, USA
| | - Donald G Phinney
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL33458, USA
| |
Collapse
|
16
|
Da Silva D, Crous A, Abrahamse H. Synergistic Effects of Photobiomodulation and Differentiation Inducers on Osteogenic Differentiation of Adipose-Derived Stem Cells in Three-Dimensional Culture. Int J Mol Sci 2024; 25:13350. [PMID: 39769115 PMCID: PMC11678880 DOI: 10.3390/ijms252413350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Osteoporosis, a common metabolic bone disorder, leads to increased fracture risk and significant morbidity, particularly in postmenopausal women and the elderly. Traditional treatments often fail to fully restore bone health and may cause side effects, prompting the exploration of regenerative therapies. Adipose-derived stem cells (ADSCs) offer potential for osteoporosis treatment, but their natural inclination toward adipogenic rather than osteogenic differentiation poses a challenge. This study investigates a novel approach combining differentiation inducers (DIs), three-dimensional (3D) hydrogel scaffolds, and photobiomodulation (PBM) to promote osteogenic differentiation of immortalised ADSCs. A dextran-based 3D hydrogel matrix, supplemented with a DI cocktail of dexamethasone, β-glycerophosphate disodium, and ascorbic acid, was used to foster osteogenesis. PBM was applied using near-infrared (825 nm), green (525 nm), and combined wavelengths at fluences of 3 J/cm2, 5 J/cm2, and 7 J/cm2 to enhance osteogenic potential. Flow cytometry identified osteoblast-specific markers, while inverted light microscopy evaluated cellular morphology. Reactive oxygen species assays measured oxidative stress, and quantitative polymerase chain reaction (qPCR) revealed upregulated gene expression linked to osteogenesis. The findings demonstrate that integrating DIs, 3D hydrogels, and PBM effectively drives osteogenic differentiation in immortalised ADSCs. The PBM enhanced osteogenic marker expression, induced morphological changes, and upregulated gene activity, presenting a promising framework for bone regeneration. Future research should assess the stability and functionality of these differentiated cells and explore their applicability in preclinical models of bone injury or degeneration. This integrative approach demonstrated specific efficacy in promoting the osteogenic differentiation of ADSCs, highlighting its potential application in developing targeted treatments for osteoporosis.
Collapse
Affiliation(s)
| | | | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa; (D.D.S.); (A.C.)
| |
Collapse
|
17
|
Osipov B, Emami AJ, Cunningham HC, Orr S, Lin YY, Jbeily EH, Punati RS, Murugesh DK, Zukowski HM, Loots GG, Carney R, Vargas D, Ferguson VL, Christiansen BA. Altered post-fracture systemic bone loss in a mouse model of osteocyte dysfunction. JBMR Plus 2024; 8:ziae135. [PMID: 39605879 PMCID: PMC11601886 DOI: 10.1093/jbmrpl/ziae135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/02/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Femur fracture leads to loss of bone at uninjured skeletal sites, which may increase risk of subsequent fracture. Osteocytes, the most abundant bone cells, can directly resorb bone matrix and regulate osteoclast and osteoblast activity, but their role in systemic bone loss after fracture remains poorly understood. In this study we used a transgenic (TG+) mouse model that overexpresses human B-cell lymphoma 2 (BCL-2) in osteoblasts and osteocytes. This causes enhanced osteoblast proliferation, followed by disruption in lacunar-canalicular connectivity and massive osteocyte death by 10 wk of age. We hypothesized that reduced viable osteocyte density would decrease the magnitude of systemic bone loss after femur fracture, reduce perilacunar remodeling, and alter callus formation. Bone remodeling was assessed using serum biomarkers of bone formation and resorption at 5 d post-fracture. We used micro-computed tomography, high resolution x-ray microscopy, mechanical testing, and Raman spectroscopy to quantify the magnitude of systemic bone loss, as well as changes in osteocyte lacunar volume, bone strength, and bone composition 2 wk post-fracture. Fracture was associated with a reduction in circulating markers of bone resorption in non-transgenic (TG-) animals. TG+ mice exhibited high bone mass in the limbs, greater cortical elastic modulus and reduced post-yield displacement. After fracture, TG+ mice lost less trabecular bone than TG- mice, but conversely TG+ mice exhibited trends toward a lower yield point and reduced femoral cortical thickness after fracture, though these were not statistically significant. Lacunar density was greater in TG+ mice, but fracture did not alter lacunar volume in TG+ or TG- mice. These findings suggest that osteocytes potentially play a significant role in the post-traumatic systemic response to fracture, though the effects differ between trabecular and cortical bone.
Collapse
Affiliation(s)
- Benjamin Osipov
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA 95817, United States
| | - Armaun J Emami
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA 95817, United States
| | - Hailey C Cunningham
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA 95817, United States
| | - Sophie Orr
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA 95817, United States
| | - Yu-Yang Lin
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA 95817, United States
| | - Elias H Jbeily
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA 95817, United States
| | - Ritvik S Punati
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA 95817, United States
| | - Deepa K Murugesh
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, United States
| | - Hannah M Zukowski
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA 95817, United States
| | - Gabriela G Loots
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA 95817, United States
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, United States
| | - Randy Carney
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, United States
| | - Diego Vargas
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Virginia L Ferguson
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Blaine A Christiansen
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA 95817, United States
| |
Collapse
|
18
|
Gupta A, Saha S, Das A, Roy Chowdhury A. Evaluating the influence on osteocyte mechanobiology within the lacunar-canalicular system for varying lacunar equancy and perilacunar elasticity: A multiscale fluid-structure interaction analysis. J Mech Behav Biomed Mater 2024; 160:106767. [PMID: 39393133 DOI: 10.1016/j.jmbbm.2024.106767] [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: 06/07/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
The lacunar morphology and perilacunar tissue properties of osteocytes in bone can vary under different physiological and pathological conditions. How these alterations collectively change the overall micromechanics of osteocytes in the lacunar-canalicular system (LCS) of an osteon still requires special focus. Therefore, a Haversian canal and LCS-based osteon model was established to evaluate the changes in the hydrodynamic environment around osteocytes under physiological loading using fluid-structure interaction analysis, followed by a sub-modelled finite element analysis to assess the mechanical responses of osteocytes and their components. Osteocytes were modelled with detailed configurations, including cytoplasm, nucleus, and cytoskeleton, and parametric variations in lacunar equancy (L.Eq) and perilacunar elasticity (Pl.E) were considered within the osteon model. The study aimed to conduct a comparative study among osteon models with varying L. Eq and Pl. E to check the resulting differences in osteocyte mechanobiology. The results demonstrated that the average mechanical stimulation of each subcellular component of osteocytes increased with decreases in L. Eq and Pl. E, reflecting conditions typically seen in young, healthy bone as per previous literature. However, hydrodynamic responses, such as fluid flow and fluid shear stress on osteocytes, varied proportionally with the elasticity difference between the bone matrix and the perilacunar region during Pl. E variation. Additionally, the findings revealed that a minimal percentage of energy was used to transmit mechanical responses through microtubules from the cell membrane to the nucleus, and this energy percentage increased with higher L. Eq. The outcomes of the study could help to quantify how the osteocyte microenvironment and its mechanosensitivity within cortical bone changes with L. Eq and Pl. E alterations in different bone conditions, from young to aged and healthy to diseased.
Collapse
Affiliation(s)
- Abhisek Gupta
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, India
| | - Subrata Saha
- Department of Restorative Dentistry, University of Washington, Seattle, WA, USA
| | - Apurba Das
- Department of Mechanical Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, India
| | - Amit Roy Chowdhury
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, India.
| |
Collapse
|
19
|
Ucci A, Giacchi L, Rucci N. Primary Bone Tumors and Breast Cancer-Induced Bone Metastases: In Vivo Animal Models and New Alternative Approaches. Biomedicines 2024; 12:2451. [PMID: 39595017 PMCID: PMC11591690 DOI: 10.3390/biomedicines12112451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Bone is the preferential site of metastasis for the most common tumors, including breast cancer. On the other hand, osteosarcoma is the primary bone cancer that most commonly occurs and causes bone cancer-related deaths in children. Several treatment strategies have been developed so far, with little or no efficacy for patient survival and with the development of side effects. Therefore, there is an urgent need to develop more effective therapies for bone primary tumors and bone metastatic disease. This almost necessarily requires the use of in vivo animal models that better mimic human pathology and at the same time follow the ethical principles for the humane use of animal testing. In this review we aim to illustrate the main and more suitable in vivo strategies employed to model bone metastases and osteosarcoma. We will also take a look at the recent technologies implemented for a partial replacement of animal testing.
Collapse
Affiliation(s)
| | | | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.U.); (L.G.)
| |
Collapse
|
20
|
Vaivads M, Pilmane M. Distribution of Immunomodulation, Protection and Regeneration Factors in Cleft-Affected Bone and Cartilage. Diagnostics (Basel) 2024; 14:2217. [PMID: 39410621 PMCID: PMC11475217 DOI: 10.3390/diagnostics14192217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Craniofacial clefts can form a significant defect within bone and cartilage, which can negatively affect tissue homeostasis and the remodeling process. Multiple proteins can affect supportive tissue growth, while also regulating local immune response and tissue protection. Some of these factors, like galectin-10 (Gal-10), nuclear factor kappa-light-chain-enhancer of activated B cells protein 65 (NF-κB p65), heat shock protein 60 (HSP60) and 70 (HSP70) and cathelicidin (LL-37), have not been well studied in cleft-affected supportive tissue, while more known tissue regeneration regulators like type I collagen (Col-I) and bone morphogenetic proteins 2 and 4 (BMP-2/4) have not been assessed jointly with immunomodulation and protective proteins. Information about the presence and interaction of these proteins in cleft-affected supportive tissue could be helpful in developing biomaterials and improving cleft treatment. METHODS Two control groups and two cleft patient groups for bone tissue and cartilage, respectively, were organized with five patients in each group. Immunohistochemistry with the semiquantitative counting method was implemented to determine Gal-10-, NF-κB p65-, HSP60-, HSP70-, LL-37-, Col-I- and BMP-2/4-positive cells within the tissue. RESULTS Factor-positive cells were identified in each study group. Multiple statistically significant correlations were identified. CONCLUSIONS A significant increase in HSP70-positive chondrocytes in cleft patients could indicate that HSP70 might be reacting to stressors caused by the local tissue defect. A significant increase in Col-I-positive osteocytes in cleft patients might indicate increased bone remodeling and osteocyte activity due to the presence of a cleft. Correlations between factors indicate notable differences in molecular interactions within each group.
Collapse
Affiliation(s)
- Mārtiņš Vaivads
- Department of Morphology, Institute of Anatomy and Anthropology, Rīga Stradiņš University, Kronvalda Boulevard 9, LV-1010 Riga, Latvia;
| | | |
Collapse
|
21
|
Spiekermann J, Höppner J, Ibnukhsein E, Sinningen K, Hanusch B, Kiewert C, Siggelkow H, Grasemann C. Description of bone health in adolescents and young persons with Klinefelter syndrome - results from a pilot study. Mol Cell Pediatr 2024; 11:9. [PMID: 39285120 PMCID: PMC11405648 DOI: 10.1186/s40348-024-00182-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/31/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND In adults with Klinefelter syndrome (KS), impaired bone health with reduced bone mineral density (BMD) has been described even in the presence of testosterone replacement therapy. The aim of the present study was to characterize bone health in young patients with KS. PATIENTS AND METHODS 20 participants aged 16.10 ± 4.28 years with KS (7 with testosterone replacement therapy) were included in the KliBONE study (DRKS 00024870). Medical history, clinical, radiographic and biochemical parameters of bone health and metabolism were obtained. Radiographic bone health index (BHI) was assessed via automated digital X-ray radiogrammetry of the left hand or via dual energy X-ray absorptiometry (DXA) of the lumbar spine and left femur in participants ≥ 16 years. Peripheral blood mononuclear cells were differentiated into osteoclasts and quantified in 7 participants and 7 healthy controls. RESULTS Mean BHI SDS was - 1.42 ± 1.22 and mean BMD z-score at the lumbar vertebrae (L1-4) was - 0.92 ± 1.00. 25-OH-vitamin D levels < 20 ng/ml were detected in 8/20. Other parameters of bone metabolism (bone-specific alkaline phosphatase, PTH, ß-crosslaps and osteocalcin) were within age-appropriate reference ranges. Serum leptin SDS was elevated (mean 2.15 ± 1.19). The number of osteoclasts in participants with KS did not differ from that of controls. CONCLUSION BHI SDS and BMD z-scores were lower than expected in young individuals with KS despite age-appropriate bone turnover markers and no apparent pathology in osteoclast differentiation. The cause of the early-onset bone phenotype requires further investigation.
Collapse
Affiliation(s)
- Julia Spiekermann
- Department of Pediatrics, Division of Rare Diseases, University Hospital of Pediatrics and Adolescent Medicine, Katholisches Klinikum Bochum, Member of ENDO ERN, Ruhr-University Bochum, Alexandrinenstraße 5, 44791, Bochum, Germany
- Center for Rare Diseases Ruhr (CeSER), Ruhr-University Bochum and Witten/Herdecke University, Bochum, Witten, Germany
| | - Jakob Höppner
- Department of Pediatrics, Division of Rare Diseases, University Hospital of Pediatrics and Adolescent Medicine, Katholisches Klinikum Bochum, Member of ENDO ERN, Ruhr-University Bochum, Alexandrinenstraße 5, 44791, Bochum, Germany
- Center for Rare Diseases Ruhr (CeSER), Ruhr-University Bochum and Witten/Herdecke University, Bochum, Witten, Germany
| | - Eliena Ibnukhsein
- Department of Pediatrics, Division of Rare Diseases, University Hospital of Pediatrics and Adolescent Medicine, Katholisches Klinikum Bochum, Member of ENDO ERN, Ruhr-University Bochum, Alexandrinenstraße 5, 44791, Bochum, Germany
| | - Kathrin Sinningen
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Beatrice Hanusch
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Cordula Kiewert
- Division of Pediatric Endocrinology, Department of Pediatrics II, Member of ENDO ERN, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Heide Siggelkow
- Clinic of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Göttingen, Germany and MVZ Endokrinologikum Göttingen, Göttingen, Germany
| | - Corinna Grasemann
- Department of Pediatrics, Division of Rare Diseases, University Hospital of Pediatrics and Adolescent Medicine, Katholisches Klinikum Bochum, Member of ENDO ERN, Ruhr-University Bochum, Alexandrinenstraße 5, 44791, Bochum, Germany.
- Center for Rare Diseases Ruhr (CeSER), Ruhr-University Bochum and Witten/Herdecke University, Bochum, Witten, Germany.
| |
Collapse
|
22
|
Giraldo-Osorno PM, Wirsig K, Asa'ad F, Omar O, Trobos M, Bernhardt A, Palmquist A. Macrophage-to-osteocyte communication: Impact in a 3D in vitro implant-associated infection model. Acta Biomater 2024; 186:141-155. [PMID: 39142531 DOI: 10.1016/j.actbio.2024.08.005] [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: 04/23/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024]
Abstract
Macrophages and osteocytes are important regulators of inflammation, osteogenesis and osteoclastogenesis. However, their interactions under adverse conditions, such as biomaterial-associated infection (BAI) are not fully understood. We aimed to elucidate how factors released from macrophages modulate osteocyte responses in an in vitro indirect 3D co-culture model. Human monocyte-derived macrophages were cultured on etched titanium disks and activated with either IL-4 cytokine (anti-inflammatory M2 phenotype) or Staphylococcus aureus secreted virulence factors to simulate BAI (pro-inflammatory M1 phenotype). Primary osteocytes in collagen gels were then stimulated with conditioned media (CM) from these macrophages. The osteocyte response was analyzed by gene expression, protein secretion, and immunostaining. M1 phenotype macrophages were confirmed by IL-1β and TNF-α secretion, and M2 macrophages by ARG-1 and MRC-1.Osteocytes receiving M1 CM revealed bone inhibitory effects, denoted by reduced secretion of bone formation osteocalcin (BGLAP) and increased secretion of the bone inhibitory sclerostin (SOST). These osteocytes also downregulated the pro-mineralization gene PHEX and upregulated the anti-mineralization gene MEPE. Additionally, exhibited pro-osteoclastic potential by upregulating pro-osteoclastic gene RANKL expression. Nonetheless, M1-stimulated osteocytes expressed a higher level of the potent pro-osteogenic factor BMP-2 in parallel with the downregulation of the bone inhibitor genes DKK1 and SOST, suggesting a compensatory feedback mechanisms. Conversely, M2-stimulated osteocytes mainly upregulated anti-osteoclastic gene OPG expression, suggesting an anti-catabolic effect. Altogether, our findings demonstrate a strong communication between M1 macrophages and osteocytes under M1 (BAI)-simulated conditions, suggesting that the BAI adverse effects on osteoblastic and osteoclastic processes in vitro are partly mediated via this communication. STATEMENT OF SIGNIFICANCE: Biomaterial-associated infections are major challenges and the underlying mechanisms in the cellular interactions are missing, especially among the major cells from the inflammatory side (macrophages as the key cell in bacterial clearance) and the regenerative side (osteocyte as main regulator of bone). We evaluated the effect of macrophage polarization driven by the stimulation with bacterial virulence factors on the osteocyte function using an indirect co-culture model, hence mimicking the scenario of a biomaterial-associated infection. The results suggest that at least part of the adverse effects of biomaterial associated infection on osteoblastic and osteoclastic processes in vitro are mediated via macrophage-to-osteocyte communication.
Collapse
Affiliation(s)
- Paula Milena Giraldo-Osorno
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Katharina Wirsig
- Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universität Dresden, Germany
| | - Farah Asa'ad
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Omar Omar
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Margarita Trobos
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anne Bernhardt
- Faculty of Medicine, Centre for Translational Bone, Joint and Soft Tissue Research, Technische Universität Dresden, Germany.
| | - Anders Palmquist
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
23
|
Karacan I, Türker KS. Exploring neuronal mechanisms of osteosarcopenia in older adults. J Physiol 2024. [PMID: 39119811 DOI: 10.1113/jp285666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Until recently, research on the pathogenesis and treatment of osteoporosis and sarcopenia has primarily focused on local and systemic humoral mechanisms, often overlooking neuronal mechanisms. However, there is a growing body of literature on the neuronal regulation of bone and skeletal muscle structure and function, which may provide insights into the pathogenesis of osteosarcopenia. This review aims to integrate these neuronal regulatory mechanisms to form a comprehensive understanding and inspire future research that could uncover novel strategies for preventing and treating osteosarcopenia. Specifically, the review explores the functional adaptation of weight-bearing bone to mechanical loading throughout evolutionary development, from Wolff's law and Frost's mechanostat theory to the mosaic hypothesis, which emphasizes neuronal regulation. The recently introduced bone osteoregulation reflex points to the importance of the osteocytic mechanoreceptive network as a receptor in this neuronal regulation mechanism. Finally, the review focuses on the bone myoregulation reflex, which is known as a mechanism by which bone loading regulates muscle functions neuronally. Considering the ageing-related regressive changes in the nerve fibres that provide both structural and functional regulation in bone and skeletal muscle tissue and the bone and muscle tissues they innervate, it is suggested that neuronal mechanisms might play a central role in explaining osteosarcopenia in older adults.
Collapse
Affiliation(s)
- Ilhan Karacan
- Physical Medicine and Rehabilitation Department, Hamidiye Medical School, Health Science University Istanbul, Istanbul, Turkey
- Istanbul Physical Therapy Rehabilitation Training and Research Hospital, Istanbul, Turkey
| | - Kemal Sıtkı Türker
- Physiology, Faculty of Dentistry, Istanbul Gelisim University, Istanbul, Turkey
| |
Collapse
|
24
|
Taskozhina G, Batyrova G, Umarova G, Issanguzhina Z, Kereyeva N. The Manganese-Bone Connection: Investigating the Role of Manganese in Bone Health. J Clin Med 2024; 13:4679. [PMID: 39200820 PMCID: PMC11355939 DOI: 10.3390/jcm13164679] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/17/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
The complex relationship between trace elements and skeletal health has received increasing attention in the scientific community. Among these minerals, manganese (Mn) has emerged as a key element affecting bone metabolism and integrity. This review examines the multifaceted role of Mn in bone health, including its effects on bone regeneration, mineralization, and overall skeletal strength. This review article is based on a synthesis of experimental models, epidemiologic studies, and clinical trials of the mechanisms of the effect of Mn on bone metabolism. Current research data show that Mn is actively involved in the processes of bone remodeling by modulating the activity of osteoblasts and osteoclasts, as well as the main cells that regulate bone formation and resorption. Mn ions have a profound effect on bone mineralization and density by intricately regulating signaling pathways and enzymatic reactions in these cells. Additionally, Mn superoxide dismutase (MnSOD), located in bone mitochondria, plays a crucial role in osteoclast differentiation and function, protecting osteoclasts from oxidative damage. Understanding the nuances of Mn's interaction with bone is essential for optimizing bone strategies, potentially preventing and managing skeletal diseases. Key findings include the stimulation of osteoblast proliferation and differentiation, the inhibition of osteoclastogenesis, and the preservation of bone mass through the RANK/RANKL/OPG pathway. These results underscore the importance of Mn in maintaining bone health and highlight the need for further research into its therapeutic potential.
Collapse
Affiliation(s)
- Gulaim Taskozhina
- Department of Laboratory Diagnostics, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Street, Aktobe 030019, Kazakhstan
| | - Gulnara Batyrova
- Department of Laboratory Diagnostics, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Street, Aktobe 030019, Kazakhstan
| | - Gulmira Umarova
- Department of Evidence-Based Medicine and Scientific Management, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Street, Aktobe 030019, Kazakhstan;
| | - Zhamilya Issanguzhina
- Department of Children Disease No. 2, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Street, Aktobe 030019, Kazakhstan;
| | - Nurgul Kereyeva
- Department of Oncology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Street, Aktobe 030019, Kazakhstan;
| |
Collapse
|
25
|
Hao Y, Yang N, Sun M, Yang S, Chen X. The role of calcium channels in osteoporosis and their therapeutic potential. Front Endocrinol (Lausanne) 2024; 15:1450328. [PMID: 39170742 PMCID: PMC11335502 DOI: 10.3389/fendo.2024.1450328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024] Open
Abstract
Osteoporosis, a systemic skeletal disorder marked by diminished bone mass and compromised bone microarchitecture, is becoming increasingly prevalent due to an aging population. The underlying pathophysiology of osteoporosis is attributed to an imbalance between osteoclast-mediated bone resorption and osteoblast-mediated bone formation. Osteoclasts play a crucial role in the development of osteoporosis through various molecular pathways, including the RANK/RANKL/OPG signaling axis, cytokines, and integrins. Notably, the calcium signaling pathway is pivotal in regulating osteoclast activation and function, influencing bone resorption activity. Disruption in calcium signaling can lead to increased osteoclast-mediated bone resorption, contributing to the progression of osteoporosis. Emerging research indicates that calcium-permeable channels on the cellular membrane play a critical role in bone metabolism by modulating these intracellular calcium pathways. Here, we provide an overview of current literature on the regulation of plasma membrane calcium channels in relation to bone metabolism with particular emphasis on their dysregulation during the progression of osteoporosis. Targeting these calcium channels may represent a potential therapeutic strategy for treating osteoporosis.
Collapse
Affiliation(s)
- Ying Hao
- College of Sports, Northwest Normal University, Lanzhou, China
| | - Ningning Yang
- College of Sports, Northwest Normal University, Lanzhou, China
| | - Mengying Sun
- College of Sports, Northwest Normal University, Lanzhou, China
| | - Shangze Yang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| |
Collapse
|
26
|
Tauer JT, Thiele T, Julien C, Ofer L, Zaslansky P, Shahar R, Willie BM. Swim training induces distinct osseous gene expression patterns in anosteocytic and osteocytic teleost fish. Bone 2024; 185:117125. [PMID: 38754573 DOI: 10.1016/j.bone.2024.117125] [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: 01/13/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
The traditional understanding of bone mechanosensation implicates osteocytes, canaliculi, and the lacunocanalicular network in biomechanical adaptation. However, recent findings challenge this notion, as shown in advanced teleost fish where anosteocytic bone lacking osteocytes are nevertheless responsive to mechanical load. To investigate specific molecular mechanisms involved in bone mechanoadaptation in osteocytic and anosteocytic fish bone, we conducted a 5-min single swim-training experiment with zebrafish and ricefish, respectively. Through RNASeq analysis of fish spines, analyzed at various time points following swim training, we uncovered distinct gene expression patterns in osteocytic and anosteocytic fish bones. Notably, osteocytic fish bone exhibited an early response to mechanical load, contrasting to a delayed response observed in anosteocytic fish bones, both within 8 h following stimulation. We identified an increase in osteoblast differentiation in anosteocytic bone following training, while chordoblast activity was delayed. This temporal response suggests a time-dependent adaptation in anosteocytic bone, indicating the presence of intricate feedback networks within bone that lacks osteocytes.
Collapse
Affiliation(s)
- Josephine T Tauer
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada; Department of Pediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Tobias Thiele
- Julius Wolff Institute and Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Catherine Julien
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Lior Ofer
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University, Rehovot, Israel
| | - Paul Zaslansky
- Department of Operative, Preventive and Pediatric Dentistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ron Shahar
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University, Rehovot, Israel
| | - Bettina M Willie
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada.
| |
Collapse
|
27
|
Hansdah K, Lui JC. Emerging Insights into the Endocrine Regulation of Bone Homeostasis by Gut Microbiome. J Endocr Soc 2024; 8:bvae117. [PMID: 38957653 PMCID: PMC11215793 DOI: 10.1210/jendso/bvae117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Indexed: 07/04/2024] Open
Abstract
Gut microbiota plays an important role in the regulation of bone homeostasis and bone health. Recent studies showed that these effects could be mediated through microbial metabolites released by the microbiota like short-chain fatty acids, metabolism of endogenous molecules such as bile acids, or a complex interplay between microbiota, the endocrine system, and the immune system. Importantly, some studies showed a reciprocal relationship between the endocrine system and gut microbiota. For instance, postmenopausal estrogen deficiency could lead to dysbiosis of the gut microbiota, which could in turn affect various immune response and bone remodeling. In addition, evidence showed that shift in the indigenous gut microbiota caused by antibiotics treatment may also impact normal skeletal growth and maturation. In this mini-review, we describe recent findings on the role of microbiome in bone homeostasis, with a particular focus on molecular mechanisms and their interactions with the endocrine and immune system. We will also discuss the recent findings on estrogen deficiency and microbiota dysbiosis, and the clinical implications for the development of new therapeutic strategies for osteoporosis and other bone disorders.
Collapse
Affiliation(s)
- Kirtal Hansdah
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Julian C Lui
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
28
|
Kim JG, Sharma AR, Lee YH, Chatterjee S, Choi YJ, Rajvansh R, Chakraborty C, Lee SS. Therapeutic Potential of Quercetin as an Antioxidant for Bone-Muscle-Tendon Regeneration and Aging. Aging Dis 2024; 16:1414-1437. [PMID: 39012676 PMCID: PMC12096925 DOI: 10.14336/ad.2024.0282] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/24/2024] [Indexed: 07/17/2024] Open
Abstract
Quercetin (QC), a naturally occurring bioflavonoid found in various fruits and vegetables, possesses many potential health benefits, primarily attributed to its robust antioxidant properties. The generation of oxidative stress in bone cells is a key modulator of their physiological behavior. Moreover, oxidative stress status influences the pathophysiology of mineralized tissues. Increasing scientific evidence demonstrates that manipulating the redox balance in bone cells might be an effective technique for developing bone disease therapies. The QC antioxidant abilities in skeletal muscle significantly enhance muscle regeneration and reduce muscle atrophy. In addition, QC has been shown to have protective effects against oxidative stress, inflammation, apoptosis, and matrix degradation in tendons, helping to maintain the structural integrity and functionality of tendons. Thus, the antioxidant properties of QC might be crucial for addressing age-related musculoskeletal disorders like osteoporosis, sarcopenia, and tendon-related inflammatory conditions. Understanding how QC influences redox signaling pathways involved in musculoskeletal disorders, including their effect on bone, muscle, and tendon differentiation, might provide insights into the diverse advantages of QC in promoting tissue regeneration and preventing cellular damage. Therefore, this study reviewed the intricate relationship among oxidative stress, inflammation, and tissue repair, affected by the antioxidative abilities of QC, in age-related musculoskeletal tissues to improve the overall health of bones, muscles, and tendons of the skeletal system. Also, reviewing the ongoing clinical trials of QC for musculoskeletal systems is encouraging. Given the positive effect of QC on musculoskeletal health, further scientific investigations and controlled human intervention studies are necessary to explore the therapeutic potential to its optimum strength.
Collapse
Affiliation(s)
- Jae Gyu Kim
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Korea.
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Korea.
| | - Yeon-Hee Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Korea.
| | - Srijan Chatterjee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Korea.
| | - Yean Jung Choi
- Department of Food and Nutrition, Sahmyook University, Seoul 01795, Korea.
| | - Roshani Rajvansh
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Korea.
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal 700126, India.
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Korea.
| |
Collapse
|
29
|
Dang AT, Ono M, Wang Z, Tosa I, Hara ES, Mikai A, Kitagawa W, Yonezawa T, Kuboki T, Oohashi T. Local E-rhBMP-2/β-TCP Application Rescues Osteocyte Dendritic Integrity and Reduces Microstructural Damage in Alveolar Bone Post-Extraction in MRONJ-like Mouse Model. Int J Mol Sci 2024; 25:6648. [PMID: 38928355 PMCID: PMC11203997 DOI: 10.3390/ijms25126648] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The pathology of medication-related osteonecrosis of the jaw (MRONJ), often associated with antiresorptive therapy, is still not fully understood. Osteocyte networks are known to play a critical role in maintaining bone homeostasis and repair, but the exact condition of these networks in MRONJ is unknown. On the other hand, the local application of E-coli-derived Recombinant Human Bone Morphogenetic Protein 2/β-Tricalcium phosphate (E-rhBMP-2/β-TCP) has been shown to promote bone regeneration and mitigate osteonecrosis in MRONJ-like mouse models, indicating its potential therapeutic application for the treatment of MRONJ. However, the detailed effect of BMP-2 treatment on restoring bone integrity, including its osteocyte network, in an MRONJ condition remains unclear. Therefore, in the present study, by applying a scanning electron microscope (SEM) analysis and a 3D osteocyte network reconstruction workflow on the alveolar bone surrounding the tooth extraction socket of an MRONJ-like mouse model, we examined the effectiveness of BMP-2/β-TCP therapy on the alleviation of MRONJ-related bone necrosis with a particular focus on the osteocyte network and alveolar bone microstructure (microcrack accumulation). The 3D osteocyte dendritic analysis showed a significant decrease in osteocyte dendritic parameters along with a delay in bone remodeling in the MRONJ group compared to the healthy counterpart. The SEM analysis also revealed a notable increase in the number of microcracks in the alveolar bone surface in the MRONJ group compared to the healthy group. In contrast, all of those parameters were restored in the E-rhBMP-2/β-TCP-treated group to levels that were almost similar to those in the healthy group. In summary, our study reveals that MRONJ induces osteocyte network degradation and microcrack accumulation, while application of E-rhBMP-2/β-TCP can restore a compromised osteocyte network and abrogate microcrack accumulation in MRONJ.
Collapse
Affiliation(s)
- Anh Tuan Dang
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan; (I.T.); (T.K.)
| | - Mitsuaki Ono
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
- Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Ziyi Wang
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
| | - Ikue Tosa
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan; (I.T.); (T.K.)
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Emilio Satoshi Hara
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan;
| | - Akihiro Mikai
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
| | - Wakana Kitagawa
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan; (I.T.); (T.K.)
| | - Tomoko Yonezawa
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan; (I.T.); (T.K.)
- Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Toshitaka Oohashi
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
| |
Collapse
|
30
|
Xu T, Yin J, Dai X, Liu T, Shi H, Zhang Y, Wang S, Yue G, Zhang Y, Zhao D, Gao S, Prentki M, Wang L, Zhang D. Cnidii Fructus: A traditional Chinese medicine herb and source of antiosteoporotic drugs. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155375. [PMID: 38507853 DOI: 10.1016/j.phymed.2024.155375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Osteoporosis (OP) is a prevalent chronic metabolic bone disease for which limited countermeasures are available. Cnidii Fructus (CF), primarily derived from Cnidium monnieri (L.) Cusson., has been tested in clinical trials of traditional Chinese medicine for the management of OP. Accumulating preclinical studies indicate that CF may be used against OP. MATERIALS AND METHODS Comprehensive documentation and analysis were conducted to retrieve CF studies related to its main phytochemical components as well as its pharmacokinetics, safety and pharmacological properties. We also retrieved information on the mode of action of CF and, in particular, preclinical and clinical studies related to bone remodeling. This search was performed from the inception of databases up to the end of 2022 and included PubMed, China National Knowledge Infrastructure, the National Science and Technology Library, the China Science and Technology Journal Database, Weipu, Wanfang, the Web of Science and the China National Patent Database. RESULTS CF contains a wide range of natural active compounds, including osthole, bergapten, imperatorin and xanthotoxin, which may underlie its beneficial effects on improving bone metabolism and quality. CF action appears to be mediated via multiple processes, including the osteoprotegerin (OPG)/receptor activator of nuclear factor-κB ligand (RANKL)/receptor activator of nuclear factor-κB (RANK), Wnt/β-catenin and bone morphogenetic protein (BMP)/Smad signaling pathways. CONCLUSION CF and its ingredients may provide novel compounds for developing anti-OP drugs.
Collapse
Affiliation(s)
- Tianshu Xu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Jiyuan Yin
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Xuan Dai
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Tianyuan Liu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Hanfen Shi
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Yueyi Zhang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Shan Wang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Gaiyue Yue
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Yanfei Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing 102488, PR China
| | - Dandan Zhao
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Sihua Gao
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China
| | - Marc Prentki
- Departments of Nutrition, Biochemistry and Molecular Medicine, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montreal, QC, H1W 4A4, Canada
| | - Lili Wang
- Department of TCM Pharmacology, Chinese Material Medica School, Beijing University of Chinese Medicine, Beijing 102488, PR China.
| | - Dongwei Zhang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, PR China.
| |
Collapse
|
31
|
Babu S, Shanmugavadivu A, Selvamurugan N. Tunable mechanical properties of chitosan-based biocomposite scaffolds for bone tissue engineering applications: A review. Int J Biol Macromol 2024; 272:132820. [PMID: 38825286 DOI: 10.1016/j.ijbiomac.2024.132820] [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: 02/20/2024] [Revised: 05/11/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
Bone tissue engineering (BTE) aims to develop implantable bone replacements for severe skeletal abnormalities that do not heal. In the field of BTE, chitosan (CS) has become a leading polysaccharide in the development of bone scaffolds. Although CS has several excellent properties, such as biodegradability, biocompatibility, and antibacterial properties, it has limitations for use in BTE because of its poor mechanical properties, increased degradation, and minimal bioactivity. To address these issues, researchers have explored other biomaterials, such as synthetic polymers, ceramics, and CS coatings on metals, to produce CS-based biocomposite scaffolds for BTE applications. These CS-based biocomposite scaffolds demonstrate superior properties, including mechanical characteristics, such as compressive strength, Young's modulus, and tensile strength. In addition, they are compatible with neighboring tissues, exhibit a controlled rate of degradation, and promote cell adhesion, proliferation, and osteoblast differentiation. This review provides a brief outline of the recent progress in making different CS-based biocomposite scaffolds and how to characterize them so that their mechanical properties can be tuned using crosslinkers for bone regeneration.
Collapse
Affiliation(s)
- Sushma Babu
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
32
|
Wang J, Xue M, Hu Y, Li J, Li Z, Wang Y. Proteomic Insights into Osteoporosis: Unraveling Diagnostic Markers of and Therapeutic Targets for the Metabolic Bone Disease. Biomolecules 2024; 14:554. [PMID: 38785961 PMCID: PMC11118602 DOI: 10.3390/biom14050554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/25/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Osteoporosis (OP), a prevalent skeletal disorder characterized by compromised bone strength and increased susceptibility to fractures, poses a significant public health concern. This review aims to provide a comprehensive analysis of the current state of research in the field, focusing on the application of proteomic techniques to elucidate diagnostic markers and therapeutic targets for OP. The integration of cutting-edge proteomic technologies has enabled the identification and quantification of proteins associated with bone metabolism, leading to a deeper understanding of the molecular mechanisms underlying OP. In this review, we systematically examine recent advancements in proteomic studies related to OP, emphasizing the identification of potential biomarkers for OP diagnosis and the discovery of novel therapeutic targets. Additionally, we discuss the challenges and future directions in the field, highlighting the potential impact of proteomic research in transforming the landscape of OP diagnosis and treatment.
Collapse
Affiliation(s)
- Jihan Wang
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China; (J.W.)
| | - Mengju Xue
- School of Medicine, Xi’an International University, Xi’an 710077, China
| | - Ya Hu
- Department of Medical College, Hunan Polytechnic of Environment and Biology, Hengyang 421000, China
| | - Jingwen Li
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China; (J.W.)
- Research and Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Zhenzhen Li
- Xi’an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China; (J.W.)
- Research and Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Yangyang Wang
- School of Electronics and Information, Northwestern Polytechnical University, Xi’an 710129, China
| |
Collapse
|
33
|
Wang Z, Chen X, Yan L, Wang W, Zheng P, Mohammadreza A, Liu Q. Antimicrobial peptides in bone regeneration: mechanism and potential. Expert Opin Biol Ther 2024; 24:285-304. [PMID: 38567503 DOI: 10.1080/14712598.2024.2337239] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Antimicrobial peptides (AMPs) are small-molecule peptides with a unique antimicrobial mechanism. Other notable biological activities of AMPs, including anti-inflammatory, angiogenesis, and bone formation effects, have recently received widespread attention. These remarkable bioactivities, combined with the unique antimicrobial mechanism of action of AMPs, have led to their increasingly important role in bone regeneration. AREAS COVERED In this review, on the one hand, we aimed to summarize information about the AMPs that are currently used for bone regeneration by reviewing published literature in the PubMed database. On the other hand, we also highlight some AMPs with potential roles in bone regeneration and their possible mechanisms of action. EXPERT OPINION The translation of AMPs to the clinic still faces many problems, but their unique antimicrobial mechanisms and other conspicuous biological activities suggest great potential. An in-depth understanding of the structure and mechanism of action of AMPs will help us to subsequently combine AMPs with different carrier systems and perform structural modifications to reduce toxicity and achieve stable release, which may be a key strategy for facilitating the translation of AMPs to the clinic.
Collapse
Affiliation(s)
- ZhiCheng Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - XiaoMan Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Liang Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - WenJie Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - PeiJia Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Atashbahar Mohammadreza
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of International Education, Southern Medical University, Guangzhou, China
| | - Qi Liu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
34
|
Ahmad M, Haffner-Luntzer M, Schoppa A, Najafova Z, Lukic T, Yorgan TA, Amling M, Schinke T, Ignatius A. Mechanical induction of osteoanabolic Wnt1 promotes osteoblast differentiation via Plat. FASEB J 2024; 38:e23489. [PMID: 38407813 DOI: 10.1096/fj.202301424rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Physical activity-induced mechanical stimuli play a crucial role in preserving bone mass and structure by promoting bone formation. While the Wnt pathway is pivotal for mediating the osteoblast response to loading, the exact mechanisms are not fully understood. Here, we found that mechanical stimulation induces osteoblastic Wnt1 expression, resulting in an upregulation of key osteogenic marker genes, including Runx2 and Sp7, while Wnt1 knockdown using siRNA prevented these effects. RNAseq analysis identified Plat as a major target through which Wnt1 exerts its osteogenic influence. This was corroborated by Plat depletion using siRNA, confirming its positive role in osteogenic differentiation. Moreover, we demonstrated that mechanical stimulation enhances Plat expression, which, in turn leads to increased expression of osteogenic markers like Runx2 and Sp7. Notably, Plat depletion by siRNA prevented this effect. We have established that Wnt1 regulates Plat expression by activating β-Catenin. Silencing Wnt1 impairs mechanically induced β-Catenin activation, subsequently reducing Plat expression. Furthermore, our findings showed that Wnt1 is essential for osteoblasts to respond to mechanical stimulation and induce Runx2 and Sp7 expression, in part through the Wnt1/β-Catenin/Plat signaling pathway. Additionally, we observed significantly reduced Wnt1 and Plat expression in bones from ovariectomy (OVX)-induced and age-related osteoporotic mouse models compared with non-OVX and young mice, respectively. Overall, our data suggested that Wnt1 and Plat play significant roles in mechanically induced osteogenesis. Their decreased expression in bones from OVX and aged mice highlights their potential involvement in post-menopausal and age-related osteoporosis, respectively.
Collapse
Affiliation(s)
- Mubashir Ahmad
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | | | - Teodora Lukic
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
35
|
Torres HM, Arnold KM, Oviedo M, Westendorf JJ, Weaver SR. Inflammatory Processes Affecting Bone Health and Repair. Curr Osteoporos Rep 2023; 21:842-853. [PMID: 37759135 PMCID: PMC10842967 DOI: 10.1007/s11914-023-00824-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review the current understanding of inflammatory processes on bone, including direct impacts of inflammatory factors on bone cells, the effect of senescence on inflamed bone, and the critical role of inflammation in bone pain and healing. RECENT FINDINGS Advances in osteoimmunology have provided new perspectives on inflammatory bone loss in recent years. Characterization of so-called inflammatory osteoclasts has revealed insights into physiological and pathological bone loss. The identification of inflammation-associated senescent markers in bone cells indicates that therapies that reduce senescent cell burden may reverse bone loss caused by inflammatory processes. Finally, novel studies have refined the role of inflammation in bone healing, including cross talk between nerves and bone cells. Except for the initial stages of fracture healing, inflammation has predominately negative effects on bone and increases fracture risk. Eliminating senescent cells, priming the osteo-immune axis in bone cells, and alleviating pro-inflammatory cytokine burden may ameliorate the negative effects of inflammation on bone.
Collapse
Affiliation(s)
- Haydee M Torres
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Katherine M Arnold
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Biomedical Engineering and Physiology Track/Regenerative Sciences Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manuela Oviedo
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
36
|
Ciaffaglione V, Rizzarelli E. Carnosine, Zinc and Copper: A Menage a Trois in Bone and Cartilage Protection. Int J Mol Sci 2023; 24:16209. [PMID: 38003398 PMCID: PMC10671046 DOI: 10.3390/ijms242216209] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Dysregulated metal homeostasis is associated with many pathological conditions, including arthritic diseases. Osteoarthritis and rheumatoid arthritis are the two most prevalent disorders that damage the joints and lead to cartilage and bone destruction. Recent studies show that the levels of zinc (Zn) and copper (Cu) are generally altered in the serum of arthritis patients. Therefore, metal dyshomeostasis may reflect the contribution of these trace elements to the disease's pathogenesis and manifestations, suggesting their potential for prognosis and treatment. Carnosine (Car) also emerged as a biomarker in arthritis and exerts protective and osteogenic effects in arthritic joints. Notably, its zinc(II) complex, polaprezinc, has been recently proposed as a drug-repurposing candidate for bone fracture healing. On these bases, this review article aims to provide an overview of the beneficial roles of Cu and Zn in bone and cartilage health and their potential application in tissue engineering. The effects of Car and polaprezinc in promoting cartilage and bone regeneration are also discussed. We hypothesize that polaprezinc could exchange Zn for Cu, present in the culture media, due to its higher sequestering ability towards Cu. However, future studies should unveil the potential contribution of Cu in the beneficial effects of polaprezinc.
Collapse
Affiliation(s)
- Valeria Ciaffaglione
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy
| | - Enrico Rizzarelli
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| |
Collapse
|
37
|
Liu Z, Wang Q, Zhang J, Qi S, Duan Y, Li C. The Mechanotransduction Signaling Pathways in the Regulation of Osteogenesis. Int J Mol Sci 2023; 24:14326. [PMID: 37762629 PMCID: PMC10532275 DOI: 10.3390/ijms241814326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Bones are constantly exposed to mechanical forces from both muscles and Earth's gravity to maintain bone homeostasis by stimulating bone formation. Mechanotransduction transforms external mechanical signals such as force, fluid flow shear, and gravity into intracellular responses to achieve force adaptation. However, the underlying molecular mechanisms on the conversion from mechanical signals into bone formation has not been completely defined yet. In the present review, we provide a comprehensive and systematic description of the mechanotransduction signaling pathways induced by mechanical stimuli during osteogenesis and address the different layers of interconnections between different signaling pathways. Further exploration of mechanotransduction would benefit patients with osteoporosis, including the aging population and postmenopausal women.
Collapse
Affiliation(s)
- Zhaoshuo Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qilin Wang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Junyou Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Sihan Qi
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yingying Duan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Chunyan Li
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing 100191, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
| |
Collapse
|
38
|
Lipreri MV, Di Pompo G, Boanini E, Graziani G, Sassoni E, Baldini N, Avnet S. Bone on-a-chip: a 3D dendritic network in a screening platform for osteocyte-targeted drugs. Biofabrication 2023; 15:045019. [PMID: 37552982 DOI: 10.1088/1758-5090/acee23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 08/08/2023] [Indexed: 08/10/2023]
Abstract
Age-related musculoskeletal disorders, including osteoporosis, are frequent and associated with long lasting morbidity, in turn significantly impacting on healthcare system sustainability. There is therefore a compelling need to develop reliable preclinical models of disease and drug screening to validate novel drugs possibly on a personalized basis, without the need ofin vivoassay. In the context of bone tissue, although the osteocyte (Oc) network is a well-recognized therapeutic target, currentin vitropreclinical models are unable to mimic its physiologically relevant and highly complex structure. To this purpose, several features are needed, including an osteomimetic extracellular matrix, dynamic perfusion, and mechanical cues (e.g. shear stress) combined with a three-dimensional (3D) culture of Oc. Here we describe, for the first time, a high throughput microfluidic platform based on 96-miniaturized chips for large-scale preclinical evaluation to predict drug efficacy. We bioengineered a commercial microfluidic device that allows real-time visualization and equipped with multi-chips by the development and injection of a highly stiff bone-like 3D matrix, made of a blend of collagen-enriched natural hydrogels loaded with hydroxyapatite nanocrystals. The microchannel, filled with the ostemimetic matrix and Oc, is subjected to passive perfusion and shear stress. We used scanning electron microscopy for preliminary material characterization. Confocal microscopy and fluorescent microbeads were used after material injection into the microchannels to detect volume changes and the distribution of cell-sized objects within the hydrogel. The formation of a 3D dendritic network of Oc was monitored by measuring cell viability, evaluating phenotyping markers (connexin43, integrin alpha V/CD51, sclerostin), quantification of dendrites, and responsiveness to an anabolic drug. The platform is expected to accelerate the development of new drug aimed at modulating the survival and function of osteocytes.
Collapse
Affiliation(s)
| | - Gemma Di Pompo
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Boanini
- Department of Chemistry 'Giacomo Ciamician', University of Bologna, Bologna, Italy
| | - Gabriela Graziani
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Enrico Sassoni
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, Bologna, Italy
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
39
|
García-Sánchez D, González-González A, Álvarez-Iglesias I, del Dujo-Gutiérrez M, Bolado-Carrancio A, Certo M, Pérez-Núñez MI, Riancho JA, Rodríguez-Rey JC, Delgado-Calle J, Pérez-Campo FM. Engineering a Pro-Osteogenic Secretome through the Transient Silencing of the Gene Encoding Secreted Frizzled Related Protein 1. Int J Mol Sci 2023; 24:12399. [PMID: 37569774 PMCID: PMC10419110 DOI: 10.3390/ijms241512399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/22/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
The evidence sustaining the regenerative properties of mesenchymal stem cells' (MSCs) secretome has prompted a paradigm change, where MSCs have shifted from being considered direct contributors to tissue regeneration toward being seen as cell factories for producing biotech medicines. We have previously designed a method to prime MSCs towards osteogenic differentiation by silencing the Wnt/β-Catenin inhibitor Sfpr1. This approach produces a significant increase in bone formation in osteoporotic mice. In this current work, we set to investigate the contribution of the secretome from the MSCs where Sfrp1 has been silenced, to the positive effect seen on bone regeneration in vivo. The conditioned media (CM) of the murine MSCs line C3H10T1/2, where Sfrp1 has been transiently silenced (CM-Sfrp1), was found to induce, in vitro, an increase in the osteogenic differentiation of this same cell line, as well as a decrease of the expression of the Wnt inhibitor Dkk1 in murine osteocytes ex vivo. A reduction in the RANKL/OPG ratio was also detected ex vivo, suggesting a negative effect of CM-Sfrp1 on osteoclastogenesis. Moreover, this CM significantly increases the mineralization of human primary MSCs isolated from osteoportotic patients in vitro. Proteomic analysis identified enrichment of proteins involved in osteogenesis within the soluble and vesicular fractions of this secretome. Altogether, we demonstrate the pro-osteogenic potential of the secretome of MSCs primmed in this fashion, suggesting that this is a valid approach to enhance the osteo-regenerative properties of MSCs' secretome.
Collapse
Affiliation(s)
- Daniel García-Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - Alberto González-González
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - Itzíar Álvarez-Iglesias
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - Mónica del Dujo-Gutiérrez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - Alfonso Bolado-Carrancio
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK;
| | - Matilde Certo
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - María Isabel Pérez-Núñez
- Department of Traumatology, Hospital Universitario Marqués de Valdecilla, University of Cantabria, 39008 Santander, Spain;
| | - José A. Riancho
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla-IDIVAL, University of Cantabria, CEBERER, 39012 Santander, Spain;
| | - José Carlos Rodríguez-Rey
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| | - Jesús Delgado-Calle
- Department of Physiology and Cell Biology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Flor María Pérez-Campo
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (I.Á.-I.); (M.d.D.-G.); (M.C.); (J.C.R.-R.)
| |
Collapse
|
40
|
Krasnova O, Neganova I. Assembling the Puzzle Pieces. Insights for in Vitro Bone Remodeling. Stem Cell Rev Rep 2023; 19:1635-1658. [PMID: 37204634 DOI: 10.1007/s12015-023-10558-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
As a highly dynamic organ, bone changes during throughout a person's life. This process is referred to as 'bone remodeling' and it involves two stages - a well-balanced osteoclastic bone resorption and an osteoblastic bone formation. Under normal physiological conditions bone remodeling is highly regulated that ensures tight coupling between bone formation and resorption, and its disruption results in a bone metabolic disorder, most commonly osteoporosis. Though osteoporosis is one of the most prevalent skeletal ailments that affect women and men aged over 40 of all races and ethnicities, currently there are few, if any safe and effective therapeutic interventions available. Developing state-of-the-art cellular systems for bone remodeling and osteoporosis can provide important insights into the cellular and molecular mechanisms involved in skeletal homeostasis and advise better therapies for patients. This review describes osteoblastogenesis and osteoclastogenesis as two vital processes for producing mature, active bone cells in the context of interactions between cells and the bone matrix. In addition, it considers current approaches in bone tissue engineering, pointing out cell sources, core factors and matrices used in scientific practice for modeling bone diseases and testing drugs. Finally, it focuses on the challenges that bone regenerative medicine is currently facing.
Collapse
Affiliation(s)
- O Krasnova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - I Neganova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia.
| |
Collapse
|
41
|
Hart DA. Regulation of Bone by Mechanical Loading, Sex Hormones, and Nerves: Integration of Such Regulatory Complexity and Implications for Bone Loss during Space Flight and Post-Menopausal Osteoporosis. Biomolecules 2023; 13:1136. [PMID: 37509172 PMCID: PMC10377148 DOI: 10.3390/biom13071136] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/04/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
During evolution, the development of bone was critical for many species to thrive and function in the boundary conditions of Earth. Furthermore, bone also became a storehouse for calcium that could be mobilized for reproductive purposes in mammals and other species. The critical nature of bone for both function and reproductive needs during evolution in the context of the boundary conditions of Earth has led to complex regulatory mechanisms that require integration for optimization of this tissue across the lifespan. Three important regulatory variables include mechanical loading, sex hormones, and innervation/neuroregulation. The importance of mechanical loading has been the target of much research as bone appears to subscribe to the "use it or lose it" paradigm. Furthermore, because of the importance of post-menopausal osteoporosis in the risk for fractures and loss of function, this aspect of bone regulation has also focused research on sex differences in bone regulation. The advent of space flight and exposure to microgravity has also led to renewed interest in this unique environment, which could not have been anticipated by evolution, to expose new insights into bone regulation. Finally, a body of evidence has also emerged indicating that the neuroregulation of bone is also central to maintaining function. However, there is still more that is needed to understand regarding how such variables are integrated across the lifespan to maintain function, particularly in a species that walks upright. This review will attempt to discuss these regulatory elements for bone integrity and propose how further study is needed to delineate the details to better understand how to improve treatments for those at risk for loss of bone integrity, such as in the post-menopausal state or during prolonged space flight.
Collapse
Affiliation(s)
- David A Hart
- Department of Surgery, Faculty of Kinesiology, and McCaig Institute for Bone & Joint Research, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
42
|
Deymier AC, Deymier PA, Latypov M, Muralidharan K. Effect of stress on the dissolution/crystallization of apatite in aqueous solution: a thermochemical equilibrium study. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2023; 381:20220242. [PMID: 37211040 DOI: 10.1098/rsta.2022.0242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/17/2022] [Indexed: 05/23/2023]
Abstract
Bone mineralization is critical to maintaining tissue mechanical function. The application of mechanical stress via exercise promotes bone mineralization via cellular mechanotransduction and increased fluid transport through the collagen matrix. However, due to its complex composition and ability to exchange ions with the surrounding body fluids, bone mineral composition and crystallization is also expected to respond to stress. Here, a combination of data from materials simulations, namely density functional theory and molecular dynamics, and experimental studies were input into an equilibrium thermodynamic model of bone apatite under stress in an aqueous solution based on the theory of thermochemical equilibrium of stressed solids. The model indicated that increasing uniaxial stress induced mineral crystallization. This was accompanied by a decrease in calcium and carbonate integration into the apatite solid. These results suggest that weight-bearing exercises can increase tissue mineralization via interactions between bone mineral and body fluid independent of cell and matrix behaviours, thus providing another mechanism by which exercise can improve bone health. This article is part of a discussion meeting issue 'Supercomputing simulations of advanced materials'.
Collapse
Affiliation(s)
- Alix C Deymier
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, USA
| | - Pierre A Deymier
- Department of Materials Science and Engineering, University of Arizona, Tucson, AZ 85721, USA
| | - Marat Latypov
- Department of Materials Science and Engineering, University of Arizona, Tucson, AZ 85721, USA
- Graduate Interdisciplinary Program in Applied Mathematics, University of Arizona, Tucson, AZ 85721, USA
| | - Krishna Muralidharan
- Department of Materials Science and Engineering, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
43
|
Matthews M, Cook E, Naguib N, Wiesner U, Lewis K. Intravital imaging of osteocyte integrin dynamic with locally injectable fluorescent nanoparticles. Bone 2023:116830. [PMID: 37327917 DOI: 10.1016/j.bone.2023.116830] [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: 03/24/2023] [Revised: 06/05/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
Osteocytes are the resident mechanosensory cells in bone. They are responsible for skeletal homeostasis and adaptation to mechanical cues. Integrin proteins play a prominent role in osteocyte mechanotransduction, but the details are not well stratified. Intravital imaging with multiphoton microscopy presents an opportunity to study molecular level mechanobiological events in vivo and presents an opportunity to study integrin dynamics in osteocytes. However, fluorescent imaging limitations with respect to excessive optical scattering and low signal to noise ratio caused by mineralized bone matrix make such investigations non-trivial. Here, we demonstrate that ultra-small and bright fluorescent core-shell silica nanoparticles (<7 nm diameter), known as Cornell Prime Dots (C'Dots), are well-suited for the in vivo bone microenvironment and can improve intravital imaging capabilities. We report validation studies for C'Dots as a novel, locally injectable in vivo osteocyte imaging tool for both non-specific cellular uptake and for targeting integrins. The pharmacokinetics of C'Dots reveal distinct sex differences in nanoparticle intracellular dynamics and clearance in osteocytes, which represents a novel topic of study in bone biology. Integrin-targeted C'Dots were used to study osteocyte integrin dynamics. To the best of our knowledge, we report here the first evidence of osteocyte integrin endocytosis and recycling in vivo. Our results provide novel insights in osteocyte biology and will open up new lines of investigation that were previously unavailable in vivo.
Collapse
Affiliation(s)
- Melia Matthews
- Department of Biomedical Engineering, Cornell University, 237 Tower Rd, Ithaca 14850, NY, USA
| | - Emily Cook
- Department of Biomedical Engineering, Cornell University, 237 Tower Rd, Ithaca 14850, NY, USA
| | - Nada Naguib
- Department of Biomedical Engineering, Cornell University, 237 Tower Rd, Ithaca 14850, NY, USA
| | - Uli Wiesner
- Department of Materials Science and Engineering, Cornell University, Bard Hall 210, Ithaca 14850, NY, USA
| | - Karl Lewis
- Department of Biomedical Engineering, Cornell University, 237 Tower Rd, Ithaca 14850, NY, USA.
| |
Collapse
|
44
|
Palumbo C, Ferretti M. Morphology and cell biology - two sides of the same coin: Importance of morphology in choosing Cre experimental models for targeting osteoblasts vs osteocytes. Bone 2023; 173:116790. [PMID: 37182755 DOI: 10.1016/j.bone.2023.116790] [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: 04/28/2023] [Accepted: 05/06/2023] [Indexed: 05/16/2023]
Abstract
The present letter to editor comments the manuscript entitled "Targeting osteocytes vs osteoblasts" by Y. Kitase and M. Prideaux, where we underlie the importance morphology in choosing Cre experimental models for targeting osteoblasts vs osteocytes.
Collapse
Affiliation(s)
- Carla Palumbo
- Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Anatomical Institutes, Via del Pozzo 71, 41124 Modena, Italy.
| | - Marzia Ferretti
- Section of Human Morphology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Anatomical Institutes, Via del Pozzo 71, 41124 Modena, Italy
| |
Collapse
|
45
|
Saul D, Menger MM, Ehnert S, Nüssler AK, Histing T, Laschke MW. Bone Healing Gone Wrong: Pathological Fracture Healing and Non-Unions-Overview of Basic and Clinical Aspects and Systematic Review of Risk Factors. Bioengineering (Basel) 2023; 10:85. [PMID: 36671657 PMCID: PMC9855128 DOI: 10.3390/bioengineering10010085] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/31/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Bone healing is a multifarious process involving mesenchymal stem cells, osteoprogenitor cells, macrophages, osteoblasts and -clasts, and chondrocytes to restore the osseous tissue. Particularly in long bones including the tibia, clavicle, humerus and femur, this process fails in 2-10% of all fractures, with devastating effects for the patient and the healthcare system. Underlying reasons for this failure are manifold, from lack of biomechanical stability to impaired biological host conditions and wound-immanent intricacies. In this review, we describe the cellular components involved in impaired bone healing and how they interfere with the delicately orchestrated processes of bone repair and formation. We subsequently outline and weigh the risk factors for the development of non-unions that have been established in the literature. Therapeutic prospects are illustrated and put into clinical perspective, before the applicability of biomarkers is finally discussed.
Collapse
Affiliation(s)
- Dominik Saul
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
- Kogod Center on Aging and Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Maximilian M. Menger
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
| | - Andreas K. Nüssler
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, 72076 Tübingen, Germany
| | - Matthias W. Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421 Homburg, Germany
| |
Collapse
|