1
|
Martiniakova M, Kovacova V, Biro R, Mondockova V, Sarocka A, Penzes N, Folwarczna J, Omelka R. Relationships among osteoporosis, redox homeostasis, and alcohol addiction: Importance of the brain-bone axis. Biomed Pharmacother 2025; 187:118063. [PMID: 40253828 DOI: 10.1016/j.biopha.2025.118063] [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: 02/21/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025] Open
Abstract
Overabundance of reactive oxygen species (oxidative distress) leads to redox homeostasis disturbance and is associated with many pathological conditions. Accumulating evidence suggests that oxidative distress may contribute to osteoporosis. This review thoroughly outlines the relationships among osteoporosis, redox homeostasis, and alcohol addiction, since these relations are not sufficiently known and subsequently summarized. The brain-bone axis plays a crucial role in alcohol-induced damage to the nervous and skeletal systems. Alterations in the nervous system can lead to osteoporosis because the central nervous system is involved in bone remodeling through various neural pathways. Conversely, as an endocrine organ, bone secretes a number of bone-derived factors (osteokines), which can influence brain function and behavior. As a result, osteoporosis is more common in individuals with neurological disorders, and sudden neurological events can rapidly increase the risk of osteoporosis. Excessive alcohol consumption is linked to many neurological complications, as well as osteoporosis, which are manifested by disrupted redox homeostasis, inflammation, neurodegeneration, inhibition of neurogenesis, decreased bone mineral density, impaired bone microarchitecture, altered mineral homeostasis, raising fracture risk, hormonal dysregulation, and altered gut microbiota composition. Compared to men, alcohol dependence has more negative consequences for women, including an increased risk of liver, cardiovascular, metabolic, mental disorders, and breast cancer. Abstinence has been demonstrated to improve bone and brain health in alcohol addiction. The discovery of the brain-bone axis may lead to the development of new therapeutic approaches for alcohol and other substance addictions. Further research is needed in this direction, as many questions remain unanswered.
Collapse
Affiliation(s)
- Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra 949 01, Slovakia.
| | - Veronika Kovacova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra 949 01, Slovakia
| | - Roman Biro
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra 949 01, Slovakia
| | - Vladimira Mondockova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra 949 01, Slovakia
| | - Anna Sarocka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra 949 01, Slovakia
| | - Noemi Penzes
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra 949 01, Slovakia
| | - Joanna Folwarczna
- Department of Pharmacology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Sosnowiec 41-200, Poland
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra 949 01, Slovakia.
| |
Collapse
|
2
|
Li XL, Zhao YQ, Miao L, An YX, Wu F, Han JY, Han JY, Tay FR, Mu Z, Jiao Y, Wang J. Strategies for promoting neurovascularization in bone regeneration. Mil Med Res 2025; 12:9. [PMID: 40025573 PMCID: PMC11874146 DOI: 10.1186/s40779-025-00596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/26/2025] [Indexed: 03/04/2025] Open
Abstract
Bone tissue relies on the intricate interplay between blood vessels and nerve fibers, both are essential for many physiological and pathological processes of the skeletal system. Blood vessels provide the necessary oxygen and nutrients to nerve and bone tissues, and remove metabolic waste. Concomitantly, nerve fibers precede blood vessels during growth, promote vascularization, and influence bone cells by secreting neurotransmitters to stimulate osteogenesis. Despite the critical roles of both components, current biomaterials generally focus on enhancing intraosseous blood vessel repair, while often neglecting the contribution of nerves. Understanding the distribution and main functions of blood vessels and nerve fibers in bone is crucial for developing effective biomaterials for bone tissue engineering. This review first explores the anatomy of intraosseous blood vessels and nerve fibers, highlighting their vital roles in bone embryonic development, metabolism, and repair. It covers innovative bone regeneration strategies directed at accelerating the intrabony neurovascular system over the past 10 years. The issues covered included material properties (stiffness, surface topography, pore structures, conductivity, and piezoelectricity) and acellular biological factors [neurotrophins, peptides, ribonucleic acids (RNAs), inorganic ions, and exosomes]. Major challenges encountered by neurovascularized materials during their clinical translation have also been highlighted. Furthermore, the review discusses future research directions and potential developments aimed at producing bone repair materials that more accurately mimic the natural healing processes of bone tissue. This review will serve as a valuable reference for researchers and clinicians in developing novel neurovascularized biomaterials and accelerating their translation into clinical practice. By bridging the gap between experimental research and practical application, these advancements have the potential to transform the treatment of bone defects and significantly improve the quality of life for patients with bone-related conditions.
Collapse
Affiliation(s)
- Xin-Ling Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Qing Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Li Miao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China
| | - Yan-Xin An
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Fan Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jin-Yu Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jing-Yuan Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Franklin R Tay
- Graduate School of Augusta University, Augusta, GA, 30912, USA
| | - Zhao Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yang Jiao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China.
| | - Jing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
3
|
El-Ghannam A, Sultana F, Dréau D, Tiwari A, Yang IH, AlFotawi R, Knabe-Ducheyne C. Novel 3D printed bioactive SiC orthopedic screw promotes bone growth associated activities by macrophages, neurons, and osteoblasts. J Biomed Mater Res A 2025; 113:e37801. [PMID: 39319410 DOI: 10.1002/jbm.a.37801] [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: 06/15/2024] [Revised: 08/26/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024]
Abstract
Ceramic additive manufacturing currently relies on binders or high-energy lasers, each with limitations affecting final product quality and suitability for medical applications. To address these challenges, our laboratory has devised a surface activation technique for ceramic particles that eliminates the necessity for polymer binders or high-energy lasers in ceramic additive manufacturing. We utilized this method to 3D print bioactive SiC orthopedic screws and evaluated their properties. The study's findings reveal that chemical oxidation of SiC activated its surface, enabling 3D printing of orthopedic screws in a binder jet printer. Post-processing impregnation with NaOH and/or NH4OH strengthened the scaffold by promoting silica crystallization or partial conversion of silicon oxide into silicon nitride. The silica surface of the SiC 3D printed orthopedic screws facilitated osteoblast and neuron adhesion and extensive axon synthesis. The silicate ions released from the 3D printed SiC screws favorably modulated macrophage immune responses toward an M1 phenotype as indicated by the inhibition of TNFα secretions and of reactive oxygen species (ROS) expression along with the promotion of IL6R shedding. In contrast, under the same experimental conditions, Ti ions released from Ti6Al4V discs promoted macrophage TNFα secretion and ROS expression. In vivo tests demonstrated direct bone deposition on the SiC scaffold and a strong interfacial bond between the implanted SiC and bone. Immunostaining showed innervation, mineralization, and vascularization of the newly formed bone at the interface with SiC. Taken altogether, the 3D printed SiC orthopedic screws foster a favorable environment for wound healing and bone regeneration. The novel 3D printing method, based on ceramic surface activation represents a significant advancement in ceramic additive manufacturing and is applicable to a wide variety of materials.
Collapse
Affiliation(s)
- Ahmed El-Ghannam
- Department of Mechanical Engineering and Engineering Science, UNC Charlotte, Charlotte, North Carolina, USA
| | - Farjana Sultana
- Department of Mechanical Engineering and Engineering Science, UNC Charlotte, Charlotte, North Carolina, USA
| | - Didier Dréau
- Department of Biological Sciences, UNC Charlotte, Charlotte, North Carolina, USA
| | - Arjun Tiwari
- Department of Mechanical Engineering and Engineering Science, UNC Charlotte, Charlotte, North Carolina, USA
| | - In Hong Yang
- Department of Mechanical Engineering and Engineering Science, UNC Charlotte, Charlotte, North Carolina, USA
| | - Randa AlFotawi
- Department of Oral and Maxillofacial Surgery, King Saud University, Riyadh, Saudi Arabia
| | | |
Collapse
|
4
|
Srivastava E, Qayoom I, Kumar A. Reduced Graphene Oxide-Substituted Nanohydroxyapatite: Rejuvenating Bone-Nerve Crosstalk with Electrical Cues in a Fragility Fracture Rat Model under Hyperglycemia. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59738-59751. [PMID: 39467155 DOI: 10.1021/acsami.4c10206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Diabetes has currently acquired the status of epidemic worldwide, and among its various pathological consequences like retinopathy and nephropathy, bone fragility fractures from diabetic osteopathy occurs in later stages and is equally destructive. Chronic hyperglycemia culminates into deteriorating microvasculature and quality of bone, making it prone to fractures. Among these, hip fractures are most common, especially in older diabetic patients apart from underlying neuropathy. Our study is an attempt to ameliorate hip fragility fracture and nerve trauma with electrical stimulation as an interface in a chronic diabetic rat model. We have fabricated reduced graphene oxide-substituted hydroxyapatite as an electroactive bone substitute and incorporated it into chitosan gelatin cryogels. The in situ reduction of graphene oxide during sintering of hydroxyapatite imparts higher potential to the fabricated composite in dealing with problem at question. The cryogels depicted optimum in vitro biocompatibility and enhanced mineralization after ectopic subcutaneous implantation in rats. The therapeutic potency of composite cryogels was evaluated in a hip fracture model with compression to the sciatic nerve in diabetic rats, mimicking the severe clinical trauma. The presence of cryogels in the femoral neck canal coupled with electrical stimulation and biochemical factors significantly improved bone regeneration in diabetic rats as depicted with microcomputed tomography analysis and histology images. The application of electrical stimulation also ameliorated the nerve trauma observed with 70% improvement in electrophysiological parameters such as the compound muscle action potential with combinatorial therapy. We therefore report the successful implication of a multitarget therapy in a chronic diabetic rat model unraveling the bone-nerve crosstalk with electroactive smart cryogels.
Collapse
Affiliation(s)
- Ekta Srivastava
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Irfan Qayoom
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
- Centre of Excellence in Orthopaedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India
- Center for Nanosciences, Indian Institute of Technology Kanpur, Kanpur 208016, India
- Centre of Excellence for Materials in Medicine, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
5
|
Shiwaku Y, Okawa H, Suzuki I, Sakai S, Egusa H, Suzuki O. Induced pluripotent stem cell-derived neural stem cells promote bone formation in mice with calvarial defects. Acta Biomater 2024; 188:93-102. [PMID: 39241820 DOI: 10.1016/j.actbio.2024.08.054] [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: 02/24/2024] [Revised: 07/27/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
Nerve-derived factors have attracted attention in bone regeneration therapy due to their ability to promote bone regeneration and nerve innervation. Mesenchymal stem cells transported to target sites promote osteogenesis. However, there are few reports on the effects of neural stem cells on bone regeneration. Therefore, the aim of this study was to investigate the role of neural stem cells in osteogenesis. Here, embryoid bodies (EB) or primary neurospheres (1NS) were generated using mouse induced pluripotent stem cells (iPS cells), which were then seeded onto gelatin (Gel) sponges. The seeded Gel sponges were then transplanted into mouse calvarial bone defects. We noted that 1NS-seeded Gel promoted bone regeneration and the presence of tartrate-resistant acid phosphatase (TRAP)-positive cells, whereas the EB-seeded Gel did not. RNA-sequencing of the 1NS-seeded and EB seeded Gels showed an upregulation of the transforming growth factor (TGF)-β signaling pathway in the 1NS-seeded Gel group. Immunostaining confirmed the presence of Id3 positive cells in mice with bone defects treated with the 1NS-seeded Gel. These findings suggest that the transplantation of neural stem cells may contribute to the promotion of bone regeneration. STATEMENT OF SIGNIFICANCE: This study aimed to investigate whether neural stem cells, when seeded in Gel sponges, promoted bone regeneration. It has been well documented that bone is tightly linked with the nervous systems. Bioscaffolds comprising factors that promote innervation and bone regeneration have been investigated for use in bone therapy. However, there is limited research on the use of neural stem cells for promoting bone formation. To assess this relationship, we conducted both in vivo and in vitro assays to determine whether neural stem cells promoted bone formation. We noted that 1NS-seeded Gel sponges promoted bone formation significantly in mice with calvarial defects after 4 weeks. This study provides a novel approach of neural stem cells for bone therapy.
Collapse
Affiliation(s)
- Yukari Shiwaku
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Hiroko Okawa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai 982-8577, Japan
| | - Susumu Sakai
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Osamu Suzuki
- Division of Craniofacial Function Engineering (Division of Biomaterials Science and Engineering), Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| |
Collapse
|
6
|
James R, Subramanyam KN, Payva F, E AP, Tv VK, Sivaramakrishnan V, Ks S. In-silico analysis predicts disruption of normal angiogenesis as a causative factor in osteoporosis pathogenesis. BMC Genom Data 2024; 25:85. [PMID: 39379846 PMCID: PMC11460074 DOI: 10.1186/s12863-024-01269-z] [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: 06/22/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
Angiogenesis-osteogenesis coupling is critical for proper functioning and maintaining the health of bones. Any disruption in this coupling, associated with aging and disease, might lead to loss of bone mass. Osteoporosis (OP) is a debilitating bone metabolic disorder that affects the microarchitecture of bones, gradually leading to fracture. Computational analysis revealed that normal angiogenesis is disrupted during the progression of OP, especially postmenopausal osteoporosis (PMOP). The genes associated with OP and PMOP were retrieved from the DisGeNET database. Hub gene analysis and molecular pathway enrichment were performed via the Cytoscape plugins STRING, MCODE, CytoHubba, ClueGO and the web-based tool Enrichr. Twenty-eight (28) hub genes were identified, eight of which were transcription factors (HIF1A, JUN, TP53, ESR1, MYC, PPARG, RUNX2 and SOX9). Analysis of SNPs associated with hub genes via the gnomAD, I-Mutant2.0, MUpro, ConSurf and COACH servers revealed the substitution F201L in IL6 as the most deleterious. The IL6 protein was modeled in the SWISS-MODEL server and the substitution was analyzed via the YASARA FoldX plugin. A positive ΔΔG (1.936) of the F201L mutant indicates that the mutated structure is less stable than the wild-type structure is. Thirteen hub genes, including IL6 and the enriched molecular pathways were found to be profoundly involved in angiogenesis/endothelial function and immune signaling. Mechanical loading of bones through weight-bearing exercises can activate osteoblasts via mechanotransduction leading to increased bone formation. The present study suggests proper mechanical loading of bone as a preventive strategy for PMOP, by which angiogenesis and the immune status of the bone can be maintained. This in silico analysis could be used to understand the molecular etiology of OP and to develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Remya James
- Department of Zoology, St. Joseph's College for Women, Alappuzha, Kerala, 688001, India.
- School of Biosciences, Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, 614043, India.
| | - Koushik Narayan Subramanyam
- Department of Orthopaedics, Sri Sathya Sai Institute of Higher Medical Sciences, Prasanthigram, Puttaparthi, Andhra Pradesh, 515134, India
| | - Febby Payva
- Department of Zoology, St. Joseph's College for Women, Alappuzha, Kerala, 688001, India
- School of Biosciences, Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, 614043, India
| | - Amrisa Pavithra E
- Department of Zoology, St. Joseph's College for Women, Alappuzha, Kerala, 688001, India
| | - Vineeth Kumar Tv
- Department of Zoology, The Cochin College, Kochi, Kerala, 682002, India.
| | - Venketesh Sivaramakrishnan
- School of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthinilayam, Puttaparthi, Andhra Pradesh, 515134, India
| | - Santhy Ks
- School of Biosciences, Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, 614043, India.
| |
Collapse
|
7
|
Xu M, Zhu M, Qin Q, Xing X, Archer M, Ramesh S, Cherief M, Li Z, Levi B, Clemens TL, James AW. Neuronal regulation of bone and tendon injury repair: a focused review. J Bone Miner Res 2024; 39:1045-1060. [PMID: 38836494 DOI: 10.1093/jbmr/zjae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 06/06/2024]
Abstract
Beyond the sensation of pain, peripheral nerves have been shown to play crucial roles in tissue regeneration and repair. As a highly innervated organ, bone can recover from injury without scar formation, making it an interesting model in which to study the role of nerves in tissue regeneration. As a comparison, tendon is a musculoskeletal tissue that is hypo-innervated, with repair often resulting in scar formation. Here, we reviewed the significance of innervation in 3 stages of injury repair (inflammatory, reparative, and remodeling) in 2 commonly injured musculoskeletal tissues: bone and tendon. Based on this focused review, we conclude that peripheral innervation is essential for phases of proper bone and tendon repair, and that nerves may dynamically regulate the repair process through interactions with the injury microenvironment via a variety of neuropeptides or neurotransmitters. A deeper understanding of neuronal regulation of musculoskeletal repair, and the crosstalk between nerves and the musculoskeletal system, will enable the development of future therapies for tissue healing.
Collapse
Affiliation(s)
- Mingxin Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Manyu Zhu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Mary Archer
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Sowmya Ramesh
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Benjamin Levi
- Department of Surgery, University of Texas Southwestern, Dallas, TX 75390, United States
| | - Thomas L Clemens
- Department of Orthopaedics, University of Maryland, Baltimore, MD 21205, United States
- Department of Research Services, Baltimore Veterans Administration Medical Center, Baltimore, MD 21201, United States
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| |
Collapse
|
8
|
Li D, Liu C, Wang H, Li Y, Wang Y, An S, Sun S. The Role of Neuromodulation and Potential Mechanism in Regulating Heterotopic Ossification. Neurochem Res 2024; 49:1628-1642. [PMID: 38416374 DOI: 10.1007/s11064-024-04118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 02/29/2024]
Abstract
Heterotopic ossification (HO) is a pathological process characterized by the aberrant formation of bone in muscles and soft tissues. It is commonly triggered by traumatic brain injury, spinal cord injury, and burns. Despite a wide range of evidence underscoring the significance of neurogenic signals in proper bone remodeling, a clear understanding of HO induced by nerve injury remains rudimentary. Recent studies suggest that injury to the nervous system can activate various signaling pathways, such as TGF-β, leading to neurogenic HO through the release of neurotrophins. These pathophysiological changes lay a robust groundwork for the prevention and treatment of HO. In this review, we collected evidence to elucidate the mechanisms underlying the pathogenesis of HO related to nerve injury, aiming to enhance our understanding of how neurological repair processes can culminate in HO.
Collapse
Affiliation(s)
- Dengju Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong First Medical University, Jinan, Shandong, China
| | - Changxing Liu
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Haojue Wang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yunfeng Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yaqi Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Senbo An
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
| | - Shui Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
9
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
10
|
Qiu J, Xu J, Cai Y, Li M, Peng Y, Xu Y, Chen G. Catgut embedding in acupoints combined with repetitive transcranial magnetic stimulation for the treatment of postmenopausal osteoporosis: study protocol for a randomized clinical trial. Front Neurol 2024; 15:1295429. [PMID: 38606276 PMCID: PMC11008468 DOI: 10.3389/fneur.2024.1295429] [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: 09/16/2023] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Background To date, the clinical modulation for bone metabolism based on the neuro-bone mass regulation theory is still not popular. The stimulation of nerve systems to explore novel treatments for Postmenopausal osteoporosis (PMOP) is urgent and significant. Preliminary research results suggested that changes brain function and structure may play a crucial role in bone metabolism with PMOP. Thus, we set up a clinical trial to investigate the effect of the combination of repetitive transcranial magnetic stimulation (rTMS) and catgut embedding in acupoints (CEA) for PMOP and to elucidate the central mechanism of this neural stimulation in regulating bone metabolism. Method This trial is a prospective and randomized controlled trial. 96 PMOP participants will be randomized in a 1:1:1 ratio into a CEA group, an rTMS group, or a combined one. Participants will receive CEA, rTMS, or combined therapy for 3 months with 8 weeks of follow-up. The primary outcomes will be the changes in Bone Mineral Density scores, total efficiency of Chinese Medicine Symptoms before and after treatment. Secondary outcomes include the McGill Pain Questionnaire Short-Form, Osteoporosis Symptom Score, Mini-Mental State Examination, and Beck Depression Inventory-II. The leptin, leptin receptor, and norepinephrine levels of peripheral blood must be measured before and after treatment. Adverse events that occur during the trial will be recorded. Discussion CEA achieves brain-bone mass regulation through the bottom-up way of peripheral-central while rTMS achieves it through the top-down stimulation of central-peripheral. CEA combined with rTMS can stimulate the peripheral-central at the same time and promote peripheral bone mass formation. The combination of CEA and rTMS may play a coordinating, synergistic, and side-effect-reducing role, which is of great clinical significance in exploring better treatment options for PMOP.Clinical trial registration: https://www.chictr.org.cn/, identifier ChiCTR2300073863.
Collapse
Affiliation(s)
- Jingjing Qiu
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - JiaZi Xu
- Clinical Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingyue Cai
- Clinical Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Minghong Li
- Clinical Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingsin Peng
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yunxiang Xu
- Clinical Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guizhen Chen
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
11
|
Ma C, Zhang Y, Cao Y, Hu CH, Zheng CX, Jin Y, Sui BD. Autonomic neural regulation in mediating the brain-bone axis: mechanisms and implications for regeneration under psychological stress. QJM 2024; 117:95-108. [PMID: 37252831 DOI: 10.1093/qjmed/hcad108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Indexed: 06/01/2023] Open
Abstract
Efficient regeneration of bone defects caused by disease or significant trauma is a major challenge in current medicine, which is particularly difficult yet significant under the emerging psychological stress in the modern society. Notably, the brain-bone axis has been proposed as a prominent new concept in recent years, among which autonomic nerves act as an essential and emerging skeletal pathophysiological factor related to psychological stress. Studies have established that sympathetic cues lead to impairment of bone homeostasis mainly through acting on mesenchymal stem cells (MSCs) and their derivatives with also affecting the hematopoietic stem cell (HSC)-lineage osteoclasts, and the autonomic neural regulation of stem cell lineages in bone is increasingly recognized to contribute to the bone degenerative disease, osteoporosis. This review summarizes the distribution characteristics of autonomic nerves in bone, introduces the regulatory effects and mechanisms of autonomic nerves on MSC and HSC lineages, and expounds the crucial role of autonomic neural regulation on bone physiology and pathology, which acts as a bridge between the brain and the bone. With the translational perspective, we further highlight the autonomic neural basis of psychological stress-induced bone loss and a series of pharmaceutical therapeutic strategies and implications toward bone regeneration. The summary of research progress in this field will add knowledge to the current landscape of inter-organ crosstalk and provide a medicinal basis for the achievement of clinical bone regeneration in the future.
Collapse
Affiliation(s)
- C Ma
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Y Zhang
- Department of Medical Rehabilitation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Y Cao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - C-H Hu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - C-X Zheng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Y Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - B-D Sui
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
12
|
Morris AJ, Parker RS, Nazzal MK, Natoli RM, Fehrenbacher JC, Kacena MA, White FA. Cracking the Code: The Role of Peripheral Nervous System Signaling in Fracture Repair. Curr Osteoporos Rep 2024; 22:193-204. [PMID: 38236511 PMCID: PMC10912155 DOI: 10.1007/s11914-023-00846-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/19/2024]
Abstract
PURPOSE OF REVIEW The traditionally understated role of neural regulation in fracture healing is gaining prominence, as recent findings underscore the peripheral nervous system's critical contribution to bone repair. Indeed, it is becoming more evident that the nervous system modulates every stage of fracture healing, from the onset of inflammation to repair and eventual remodeling. RECENT FINDINGS Essential to this process are neurotrophins and neuropeptides, such as substance P, calcitonin gene-related peptide, and neuropeptide Y. These molecules fulfill key roles in promoting osteogenesis, influencing inflammation, and mediating pain. The sympathetic nervous system also plays an important role in the healing process: while local sympathectomies may improve fracture healing, systemic sympathetic denervation impairs fracture healing. Furthermore, chronic activation of the sympathetic nervous system, often triggered by stress, is a potential impediment to effective fracture healing, marking an important area for further investigation. The potential to manipulate aspects of the nervous system offers promising therapeutic possibilities for improving outcomes in fracture healing. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
Collapse
Affiliation(s)
- Ashlyn J Morris
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Reginald S Parker
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Murad K Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roman M Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jill C Fehrenbacher
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| | - Fletcher A White
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
13
|
Cherief M, Xu J, Li Z, Tower RJ, Ramesh S, Qin Q, Gomez-Salazar M, Yea JH, Lee S, Negri S, Xu M, Price T, Kendal AR, Fan CM, Clemens TL, Levi B, James AW. TrkA-mediated sensory innervation of injured mouse tendon supports tendon sheath progenitor cell expansion and tendon repair. Sci Transl Med 2023; 15:eade4619. [PMID: 38117901 DOI: 10.1126/scitranslmed.ade4619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 11/29/2023] [Indexed: 12/22/2023]
Abstract
Peripheral neurons terminate at the surface of tendons partly to relay nociceptive pain signals; however, the role of peripheral nerves in tendon injury and repair remains unclear. Here, we show that after Achilles tendon injury in mice, there is new nerve growth near tendon cells that express nerve growth factor (NGF). Conditional deletion of the Ngf gene in either myeloid or mesenchymal mouse cells limited both innervation and tendon repair. Similarly, inhibition of the NGF receptor tropomyosin receptor kinase A (TrkA) abrogated tendon healing in mouse tendon injury. Sural nerve transection blocked the postinjury increase in tendon sensory innervation and the expansion of tendon sheath progenitor cells (TSPCs) expressing tubulin polymerization promoting protein family member 3. Single cell and spatial transcriptomics revealed that disruption of sensory innervation resulted in dysregulated inflammatory signaling and transforming growth factor-β (TGFβ) signaling in injured mouse tendon. Culture of mouse TSPCs with conditioned medium from dorsal root ganglia neuron further supported a role for neuronal mediators and TGFβ signaling in TSPC proliferation. Transcriptomic and histologic analyses of injured human tendon biopsy samples supported a role for innervation and TGFβ signaling in human tendon regeneration. Last, treating mice after tendon injury systemically with a small-molecule partial agonist of TrkA increased neurovascular response, TGFβ signaling, TSPC expansion, and tendon tissue repair. Although further studies should investigate the potential effects of denervation on mechanical loading of tendon, our results suggest that peripheral innervation is critical for the regenerative response after acute tendon injury.
Collapse
Affiliation(s)
- Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Robert J Tower
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Sowmya Ramesh
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Ji-Hye Yea
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Seungyong Lee
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Orthopaedics and Traumatology, University of Verona, Verona 37129, Italy
| | - Mingxin Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Theodore Price
- Department of Neuroscience, Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, TX 75080, USA
| | - Adrian R Kendal
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, Windmill Road, Oxford OX3 7LD, UK
| | - Chen-Ming Fan
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21208, USA
| | - Thomas L Clemens
- Department of Orthopaedics, University of Maryland, Baltimore, MD 21205, USA
- Baltimore Veterans Administration Medical Center, Baltimore, MD 21201, USA
| | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
14
|
Wang J, Yang Q, Saiding Q, Chen L, Liu M, Wang Z, Xiang L, Deng L, Chen Y, Cui W. Geometric Angles and Gene Expression in Cells for Structural Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304111. [PMID: 37775309 PMCID: PMC10646237 DOI: 10.1002/advs.202304111] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/18/2023] [Indexed: 10/01/2023]
Abstract
Geometry and angles play crucial roles in cellular processes; however, its mechanisms of regulation remain unclear. In this study, a series of three dimensional (3D)-printed microfibers with different geometries is constructed using a near-field electrostatic printing technique to investigate the regulatory mechanisms of geometry on stem cell function and bone regeneration. The scaffolds precisely mimicked cell dimensions with high porosity and interoperability. Compared with other spatial topography angles, microfibers with a 90° topology can significantly promote the expression of osteogenic gene proteins in bone marrow-derived mesenchymal stem cells (BMSCs). The effects of different spatial structures on the expression profiles of BMSCs differentiation genes are correlated and validated using microRNA sequencing. Enrichment analysis shows that the 90° microfibers promoted osteogenesis in BMSCs by significantly upregulating miR-222-5p/cbfb/Runx2 expression. The ability of the geometric architecture to promote bone regeneration, as assessed using the cranial defect model, demonstrates that the 90° fiber scaffolds significantly promote new bone regeneration and neovascular neural network formation. This study is the first to elucidate the relationship between angular geometry and cellular gene expression, contributing significantly to the understanding of how geometric architecture can promote stem cell differentiation, proliferation, and function for structural bone regeneration.
Collapse
Affiliation(s)
- Juan Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Qianhao Yang
- Department of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
| | - Qimanguli Saiding
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Liang Chen
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Mingyue Liu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Zhen Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Lei Xiang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Lianfu Deng
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yixuan Chen
- Department of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| |
Collapse
|
15
|
Xiao Y, Han C, Wang Y, Zhang X, Bao R, Li Y, Chen H, Hu B, Liu S. Interoceptive regulation of skeletal tissue homeostasis and repair. Bone Res 2023; 11:48. [PMID: 37669953 PMCID: PMC10480189 DOI: 10.1038/s41413-023-00285-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 05/08/2023] [Accepted: 06/22/2023] [Indexed: 09/07/2023] Open
Abstract
Recent studies have determined that the nervous system can sense and respond to signals from skeletal tissue, a process known as skeletal interoception, which is crucial for maintaining bone homeostasis. The hypothalamus, located in the central nervous system (CNS), plays a key role in processing interoceptive signals and regulating bone homeostasis through the autonomic nervous system, neuropeptide release, and neuroendocrine mechanisms. These mechanisms control the differentiation of mesenchymal stem cells into osteoblasts (OBs), the activation of osteoclasts (OCs), and the functional activities of bone cells. Sensory nerves extensively innervate skeletal tissues, facilitating the transmission of interoceptive signals to the CNS. This review provides a comprehensive overview of current research on the generation and coordination of skeletal interoceptive signals by the CNS to maintain bone homeostasis and their potential role in pathological conditions. The findings expand our understanding of intersystem communication in bone biology and may have implications for developing novel therapeutic strategies for bone diseases.
Collapse
Affiliation(s)
- Yao Xiao
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China
| | - Changhao Han
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China
| | - Yunhao Wang
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Xinshu Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China
| | - Rong Bao
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China
| | - Yuange Li
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China
| | - Huajiang Chen
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Bo Hu
- Spine Center, Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| | - Shen Liu
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, PR China.
| |
Collapse
|
16
|
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: 2] [Impact Index Per Article: 1.0] [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
|
17
|
Skedros JG, Cronin JT, Dayton MR, Bloebaum RD, Bachus KN. Exploration of the synergistic role of cortical thickness asymmetry ("Trabecular Eccentricity" concept) in reducing fracture risk in the human femoral neck and a control bone (Artiodactyl Calcaneus). J Theor Biol 2023; 567:111495. [PMID: 37068584 DOI: 10.1016/j.jtbi.2023.111495] [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: 01/19/2023] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 04/19/2023]
Abstract
The mechanobiology of the human femoral neck is a focus of research for many reasons including studies that aim to curb age-related bone loss that contributes to a near-exponential rate of hip fractures. Many believe that the femoral neck is often loaded in rather simple bending, which causes net tension stress in the upper (superior) femoral neck and net compression stress in its inferior aspect ("T/C paradigm"). This T/C loading regime lacks in vivo proof. The "C/C paradigm" is a plausible alternative simplified load history that is characterized by a gradient of net compression across the entire femoral neck; action of the gluteus medius and external rotators of the hip are important in this context. It is unclear which paradigm is at play in natural loading due to lack of in vivo bone strain data and deficiencies in understanding mechanisms and manifestations of bone adaptation in tension vs. compression. For these reasons, studies of the femoral neck would benefit from being compared to a 'control bone' that has been proven, by strain data, to be habitually loaded in bending. The artiodactyl (sheep and deer) calcaneus model has been shown to be a very suitable control in this context. However, the application of this control in understanding the load history of the femoral neck has only been attempted in two prior studies, which did not examine the interplay between cortical and trabecular bone, or potential load-sharing influences of tendons and ligaments. Our first goal is to compare fracture risk factors of the femoral neck in both paradigms. Our second goal is to compare and contrast the deer calcaneus to the human femoral neck in terms of fracture risk factors in the T/C paradigm (the C/C paradigm is not applicable in the artiodactyl calcaneus due to its highly constrained loading). Our third goal explores interplay between dorsal/compression and plantar/tension regions of the deer calcaneus and the load-sharing roles of a nearby ligament and tendon, with insights for translation to the femoral neck. These goals were achieved by employing the analytical model of Fox and Keaveny (J. Theoretical Biology 2001, 2003) that estimates fracture risk factors of the femoral neck. This model focuses on biomechanical advantages of the asymmetric distribution of cortical bone in the direction of habitual loading. The cortical thickness asymmetry of the femoral neck (thin superior cortex, thick inferior cortex) reflects the superior-inferior placement of trabecular bone (i.e., "trabecular eccentricity," TE). TE helps the femoral neck adapt to typical stresses and strains through load-sharing between superior and inferior cortices. Our goals were evaluated in the context of TE. Results showed the C/C paradigm has lower risk factors for the superior cortex and for the overall femoral neck, which is clinically relevant. TE analyses of the deer calcaneus revealed important synergism in load-sharing between the plantar/tension cortex and adjacent ligament/tendon, which challenges conventional understanding of how this control bone achieves functional adaptation. Comparisons with the control bone also exposed important deficiencies in current understanding of human femoral neck loading and its potential histocompositional adaptations.
Collapse
Affiliation(s)
- John G Skedros
- University of Utah, Department of Orthopaedics, Salt Lake City, UT, USA; Research Service, Veterans Affairs Medical Center, Salt Lake City, UT, USA.
| | - John T Cronin
- University of Utah, Department of Orthopaedics, Salt Lake City, UT, USA
| | - Michael R Dayton
- University of Colorado, Department of Orthopedics, Aurora, CO, USA
| | - Roy D Bloebaum
- University of Utah, Department of Orthopaedics, Salt Lake City, UT, USA; Research Service, Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Kent N Bachus
- University of Utah, Department of Orthopaedics, Salt Lake City, UT, USA; Research Service, Veterans Affairs Medical Center, Salt Lake City, UT, USA
| |
Collapse
|
18
|
Jiang Y, Zhu Z, Wang B, Yuan Y, Zhang Q, Li Y, Du Y, Gong P. Neuronal TRPV1-CGRP axis regulates bone defect repair through Hippo signaling pathway. Cell Signal 2023:110779. [PMID: 37336315 DOI: 10.1016/j.cellsig.2023.110779] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Transient receptor potential vanilloid type 1 (TRPV1) is highly expressed on sensory neurons where it serves as a polymodal receptor for detecting physical and chemical stimuli. However, the role of TRPV1 in bone metabolism remains largely unclear. This study aimed to investigate the underlying mechanism of neuronal TRPV1 in regulating bone defect repair. In vivo experiment verified that TRPV1 activation could trigger dorsal root ganglion (DRG) producing the neuropeptide calcitonin gene-related peptide (CGRP) in mice. The accelerated bone healing of femoral defect in this process was observed compared to the control group (p < 0.05). Conversely, Trpv1 knockdown led to the reduced CGRP expression in DRG and nerves innervating femur bone tissue, following impaired bone formation and osteogenic capability in the defect region (p < 0.05), which could be rescued by local CGRP treatment. In vitro, results revealed that TRPV1 function in DRG neurons contributed essentially to the regulation of osteoblast physiology through affecting the production and secretion of CGRP. The capsaicin-activated neuronal TRPV1-CGRP axis could enhance the proliferation, migration and differentiation of osteoblasts (p < 0.05). Furthermore, we found that the promoting role of neuronal TRPV1 in osteogenesis were associated with Hippo signaling pathway, reflected by the phosphorylation protein level of large tumor suppressor 1 (LATS1), MOB kinase activator 1 (MOB1) and Yes-associated protein (YAP), as well as the subcellular location of YAP. Our study clarified the effects and intrinsic mechanisms of neuronal TRPV1 on bone defect repair, which might offer us a therapeutic implication for bone disorders.
Collapse
Affiliation(s)
- Yixuan Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhanfeng Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ying Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanxi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Du
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
19
|
Penolazzi L, Straudi S, Lamberti N, Lambertini E, Bianchini C, Manfredini F, Piva R. Clinically-driven design of novel methods of investigation on skeletal health status in neurological disorders. The case of the traumatic brain injuries. Front Neurol 2023; 14:1176420. [PMID: 37265470 PMCID: PMC10230040 DOI: 10.3389/fneur.2023.1176420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/24/2023] [Indexed: 06/03/2023] Open
|
20
|
Zhang Z, Wang F, Huang X, Sun H, Xu J, Qu H, Yan X, Shi W, Teng W, Jin X, Shao Z, Zhang Y, Zhao S, Wu Y, Ye Z, Yu X. Engineered Sensory Nerve Guides Self-Adaptive Bone Healing via NGF-TrkA Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206155. [PMID: 36725311 PMCID: PMC10074090 DOI: 10.1002/advs.202206155] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/10/2023] [Indexed: 06/17/2023]
Abstract
The upstream role of sensory innervation during bone homeostasis is widely underestimated in bone repairing strategies. Herein, a neuromodulation approach is proposed to orchestrate bone defect healing by constructing engineered sensory nerves (eSN) in situ to leverage the adaptation feature of SN during tissue formation. NGF liberated from ECM-constructed eSN effectively promotes sensory neuron differentiation and enhances CGRP secretion, which lead to improved RAOECs mobility and osteogenic differentiation of BMSC. In turn, such eSN effectively drives ossification in vivo via NGF-TrkA signaling pathway, which substantially accelerates critical size bone defect healing. More importantly, eSN also adaptively suppresses excessive bone formation and promotes bone remodeling by activating osteoclasts via CGRP-dependent mechanism when combined with BMP-2 delivery, which ingeniously alleviates side effects of BMP-2. In sum, this eSN approach offers a valuable avenue to harness the adaptive role of neural system to optimize bone homeostasis under various clinical scenario.
Collapse
Affiliation(s)
- Zengjie Zhang
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Fangqian Wang
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Xin Huang
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Hangxiang Sun
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Jianxiang Xu
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Hao Qu
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Xiaobo Yan
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Wei Shi
- Department of OrthopedicTaizhou First People's HospitalWenzhou Medical University218 Hengjie Road, Huangyan DistrictTaizhou CityZhejiang Province318020P. R. China
| | - Wangsiyuan Teng
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Xiaoqiang Jin
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Zhenxuan Shao
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Yongxing Zhang
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Shenzhi Zhao
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Yan Wu
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Zhaoming Ye
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| | - Xiaohua Yu
- Department of Orthopedic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineOrthopedics Research Institute of Zhejiang UniversityKey Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province88 Jiefang RoadHangzhou CityZhejiang Province310003P. R. China
| |
Collapse
|
21
|
Asada N, Katayama Y. A mysterious triangle of blood, bones, and nerves. J Bone Miner Metab 2023; 41:404-414. [PMID: 36752904 DOI: 10.1007/s00774-023-01402-5] [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: 09/29/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023]
Abstract
The relationship between bone tissue and bone marrow, which is responsible for hematopoiesis, is inseparable. Osteoblasts and osteocytes, which produce and consist of bone tissue, regulate the function of hematopoietic stem cells (HSC), the ancestors of all hematopoietic cells in the bone marrow. The peripheral nervous system finely regulates bone remodeling in bone tissue and modulates HSC function within the bone marrow, either directly or indirectly via modification of the HSC niche function. Peripheral nerve signals also play an important role in the development and progression of malignant tumors (including hematopoietic tumors) and normal tissues, and peripheral nerve control is emerging as a potential new therapeutic target. In this review, we summarize recent findings on the linkage among blood system, bone tissue, and peripheral nerves.
Collapse
Affiliation(s)
- Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Yoshio Katayama
- Division of Hematology, Department of Medicine, Kobe University Hospital, 7-5-2 Kusunoki-Cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
22
|
Dharnipragada R, Ahiarakwe U, Gupta R, Abdilahi A, Butterfield J, Naik A, Parr A, Morse LR. Pharmacologic and nonpharmacologic treatment modalities for bone loss in SCI - Proposal for combined approach. J Clin Densitom 2023; 26:101359. [PMID: 36931948 DOI: 10.1016/j.jocd.2023.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023]
Abstract
Increased risk of bone fracture due to bone mineral density (BMD) loss is a serious consequence of spinal cord injury (SCI). Traditionally, pharmaceutical approaches, such as bisphosphonates, have been prescribed to prevent bone loss. However, there is controversy in the literature regarding efficacy of these medications to mitigate the drastic bone loss following SCI. Individuals with SCI are particularly at risk of osteoporosis because of the lack of ambulation and weight bearing activities. In the past two decades, functional electric stimulation (FES) has allowed for another approach to treat bone loss. FES approaches are expanding into various modalities such as cycling and rowing exercises and show promising outcomes with minimal consequences. In addition, these non-pharmacological treatments can elevate overall physical and mental health. This article provides an overview of efficacy of different treatment options for BMD loss for SCI and advocates for a combined approach be pursued in standard of care.
Collapse
Affiliation(s)
- Rajiv Dharnipragada
- University of Minnesota Medical School, Twin-Cities, Minneapolis, MN 55455, USA
| | | | - Ribhav Gupta
- University of Minnesota Medical School, Twin-Cities, Minneapolis, MN 55455, USA; Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Abdiasis Abdilahi
- University of Minnesota Medical School, Twin-Cities, Minneapolis, MN 55455, USA
| | - Jack Butterfield
- University of Minnesota Medical School, Twin-Cities, Minneapolis, MN 55455, USA
| | - Anant Naik
- Carle Illinois College of Medicine, University of Illinois Urbana Champaign, Champaign IL, 61801, USA
| | - Ann Parr
- Department of Neurosurgery, University of Minnesota Twin-Cities, Minneapolis, MN 55455, USA
| | - Leslie R Morse
- Department of Rehabilitation Medicine, University of Minnesota Twin-Cities, Minneapolis, MN 55455, USA.
| |
Collapse
|
23
|
Pappalardo XG, Testa G, Pellitteri R, Dell’Albani P, Rodolico M, Pavone V, Parano E. Early Life Stress (ELS) Effects on Fetal and Adult Bone Development. CHILDREN (BASEL, SWITZERLAND) 2023; 10:102. [PMID: 36670652 PMCID: PMC9856960 DOI: 10.3390/children10010102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/21/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023]
Abstract
Early life stress (ELS) refers to harmful environmental events (i.e., poor maternal health, metabolic restraint, childhood trauma) occurring during the prenatal and/or postnatal period, which may cause the 'epigenetic corruption' of cellular and molecular signaling of mental and physical development. While the impact of ELS in a wide range of human diseases has been confirmed, the ELS susceptibility to bone diseases has been poorly explored. In this review, to understand the potential mediating pathways of ELS in bone diseases, PRISMA criteria were used to analyze different stress protocols in mammal models and the effects elicited in dams and their progeny. Data collected, despite the methodological heterogeneity, show that ELS interferes with fetal bone formation, also revealing that the stress type and affected developmental phase may influence the variety and severity of bone anomalies. Interestingly, these findings highlight the maternal and fetal ability to buffer stress, establishing a new role for the placenta in minimizing ELS perturbations. The functional link between ELS and bone impairments will boost future investigations on maternal stress transmission to the fetus and, parallelly, help the assessment of catch-up mechanisms of skeleton adaptations from the cascading ELS effects.
Collapse
Affiliation(s)
- Xena Giada Pappalardo
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy
| | - Gianluca Testa
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, Hospital Policlinico “Rodolico-San Marco”, University of Catania, 95123 Catania, Italy
| | - Rosalia Pellitteri
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy
| | - Paola Dell’Albani
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy
| | - Margherita Rodolico
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy
| | - Vito Pavone
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, Hospital Policlinico “Rodolico-San Marco”, University of Catania, 95123 Catania, Italy
| | - Enrico Parano
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy
| |
Collapse
|
24
|
Calcitonin Gene-Related Peptide Is Potential Therapeutic Target OF Osteoporosis. Heliyon 2022; 8:e12288. [DOI: 10.1016/j.heliyon.2022.e12288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/31/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
|
25
|
Miao KZ, Cozzone A, Caetano-Lopes J, Harris MP, Fisher S. Osteoclast activity sculpts craniofacial form to permit sensorineural patterning in the zebrafish skull. Front Endocrinol (Lausanne) 2022; 13:969481. [PMID: 36387889 PMCID: PMC9664155 DOI: 10.3389/fendo.2022.969481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
Efforts to understand the morphogenesis of complex craniofacial structures have largely focused on the role of chondrocytes and osteoblasts. Along with these bone-creating cells, bone-resorbing osteoclasts are critical in homeostasis of adult skeletal structures, but there is currently limited information on their role in the complex morphogenetic events of craniofacial development. Fundamental aspects of skull formation and general skeletal development are conserved from zebrafish to mammals. Using a cathepsinK reporter, we documented osteoclast location in the developing zebrafish skull over several weeks, from 5.18 mm to 9.6 mm standard length (approximately 15 to 34 days post fertilization). While broad distribution of osteoclasts is consistent across individuals, they are sparse and the exact locations vary among fish and across developmental time points. Interestingly, we observed osteoclasts concentrating at areas associated with neuromasts and their associated nerves, in particular the hyomandibular foramina and around the supraorbital lateral line. These are areas of active remodeling. In contrast, other areas of rapid bone growth, such as the osteogenic fronts of the frontal and parietal bones, show no particular concentration of osteoclasts, suggesting that they play a special role in shaping bone near neuromasts and nerves. In csf1ra mutants lacking functional osteoclasts, the morphology of the cranial bone was disrupted in both areas. The hyomandibular foramen is present in the initial cartilage template, but after the initiation of ossification, the diameter of the canal is significantly smaller in the absence of osteoclasts. The diameter of the supraorbital lateral line canals was also reduced in the mutants, as was the number of pores associated with neuromasts, which allow for the passage of associated nerves through the bone. Our findings define important and previously unappreciated roles for osteoclast activity in shaping craniofacial skeletal structures with a particular role in bone modeling around peripheral cranial nerves, providing a scaffold for wiring the sensioneural system during craniofacial development. This has important implications for the formation of the evolutionarily diverse lateral line system, as well understanding the mechanism of neurologic sequelae of congenital osteoclast dysfunction in human craniofacial development.
Collapse
Affiliation(s)
- Kelly Z. Miao
- Department of Pharmacology and Experimental Therapeutics, Boston University Aram V. Chobanian & Edward Avedisian School of Medicine, Boston, MA, United States
| | - Austin Cozzone
- Department of Pharmacology and Experimental Therapeutics, Boston University Aram V. Chobanian & Edward Avedisian School of Medicine, Boston, MA, United States
| | - Joana Caetano-Lopes
- Department of Orthopaedic Surgery, Boston Children’s Hospital, Boston, MA, United States
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - Matthew P. Harris
- Department of Orthopaedic Surgery, Boston Children’s Hospital, Boston, MA, United States
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - Shannon Fisher
- Department of Pharmacology and Experimental Therapeutics, Boston University Aram V. Chobanian & Edward Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
26
|
Şen S, Erber R. Neuronal Guidance Molecules in Bone Remodeling and Orthodontic Tooth Movement. Int J Mol Sci 2022; 23:ijms231710077. [PMID: 36077474 PMCID: PMC9456342 DOI: 10.3390/ijms231710077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
During orthodontic tooth movement, mechanically induced remodeling occurs in the alveolar bone due to the action of orthodontic forces. The number of factors identified to be involved in mechanically induced bone remodeling is growing steadily. With the uncovering of the functions of neuronal guidance molecules (NGMs) for skeletal development as well as for bone homeostasis, NGMs are now also among the potentially significant factors for the regulation of bone remodeling during orthodontic tooth movement. This narrative review attempts to summarize the functions of NGMs in bone homeostasis and provides insight into the currently sparse literature on the functions of these molecules during orthodontic tooth movement. Presently, four families of NGMs are known: Netrins, Slits, Semaphorins, ephrins and Eph receptors. A search of electronic databases revealed roles in bone homeostasis for representatives from all four NGM families. Functions during orthodontic tooth movement, however, were only identified for Semaphorins, ephrins and Eph receptors. For these, crucial prerequisites for participation in the regulation of orthodontically induced bone remodeling, such as expression in cells of the periodontal ligament and in the alveolar bone, as well as mechanical inducibility, were shown, which suggests that the importance of NGMs in orthodontic tooth movement may be underappreciated to date and further research might be warranted.
Collapse
Affiliation(s)
- Sinan Şen
- Department of Orthodontics, University Medical Center Schleswig-Holstein, Campus Kiel, Christian Albrechts University, 24105 Kiel, Germany
- Correspondence: ; Tel.: +49-431-5002-6301
| | - Ralf Erber
- Department of Orthodontics and Dentofacial Orthopedics, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| |
Collapse
|
27
|
Hu Y, Cui J, Liu H, Wang S, Zhou Q, Zhang H, Guo J, Cao L, Chen X, Xu K, Su J. Single-cell RNA-sequencing analysis reveals the molecular mechanism of subchondral bone cell heterogeneity in the development of osteoarthritis. RMD Open 2022. [PMCID: PMC9462384 DOI: 10.1136/rmdopen-2022-002314] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The cellular composition and underlying spatiotemporal transformation processes of subchondral bone in osteoarthritis (OA) remain unknown. Herein, various cell subsets from tibial plateau of patients with OA are identified, and the mechanism of subchondral microstructure alteration is elaborated using single-cell RNA sequencing technique. We identified two novel endothelial cell (EC) populations characterised by either exosome synthesis and inflammation response or vascular function and angiogenesis. Three osteoblast (OB) subtypes are introduced, separately related to vascularisation, matrix manufacturing and matrix mineralisation. The distinct roles and functions of these novel phenotypes in OA development are further discussed as well as interaction network between these subpopulations. The variation tendency of each population is testified in a destabilisation of the medial meniscus mouse model. The identification of cell types demonstrates a novel taxonomy and mechanism for ECs and OBs inside subchondral bone area provides new insights into the physiological and pathological behaviours of subchondral bone in OA pathogenesis.
Collapse
Affiliation(s)
- Yan Hu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Jin Cui
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Sicheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Qirong Zhou
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| | - Hao Zhang
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| | - Jiawei Guo
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| | - Liehu Cao
- Department of Orthopedics, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Xiao Chen
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| |
Collapse
|
28
|
Kim NR, David K, Sommers V, Schollaert D, Deboel L, Ohlsson C, Gustafsson JÅ, Antonio L, Decallonne B, Claessens F, Vanderschueren D, Dubois V. Inactivation of AR or ERα in Extrahypothalamic Neurons Does not Affect Osteogenic Response to Loading in Male Mice. Endocrinology 2022; 163:6594680. [PMID: 35640239 DOI: 10.1210/endocr/bqac080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Indexed: 11/19/2022]
Abstract
Failure of bone mass maintenance in spite of functional loading is an important contributor to osteoporosis and related fractures. While the link between sex steroids and the osteogenic response to loading is well established, the underlying mechanisms are unknown, hampering clinical relevance. Androgens inhibit mechanoresponsiveness in male mice, but the cell type mediating this effect remains unidentified. To evaluate the role of neuronal sex steroid receptor signaling in the male bone's adaptive capacity, we subjected adult male mice with an extrahypothalamic neuron-specific knockout of the androgen receptor (N-ARKO) or the estrogen receptor alpha (N-ERαKO) to in vivo mechanical stimulation of the tibia. Loading increased cortical thickness in the control animals mainly through periosteal expansion, as total cross-sectional tissue area and cortical bone area but not medullary area were higher in the loaded than the unloaded tibia. Trabecular bone volume fraction also increased upon loading in the control group, mostly due to trabecular thickening. N-ARKO and N-ERαKO males displayed a loading response at both the cortical and trabecular bone compartments that was not different from their control littermates. In conclusion, we show that the presence of androgen receptor or estrogen receptor alpha in extrahypothalamic neurons is dispensable for the osteogenic response to mechanical loading in male mice.
Collapse
Affiliation(s)
- Na Ri Kim
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Karel David
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Vera Sommers
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Dieter Schollaert
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Ludo Deboel
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204-5056, USA
| | - Leen Antonio
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Vanessa Dubois
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
- Basic and Translational Endocrinology, Department of Basic and Applied Medical Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
29
|
Kim JH, Kim K, Kim I, Seong S, Koh JT, Kim N. Overexpression of Neurogenin 1 Negatively Regulates Osteoclast and Osteoblast Differentiation. Int J Mol Sci 2022; 23:ijms23126708. [PMID: 35743149 PMCID: PMC9223505 DOI: 10.3390/ijms23126708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/05/2023] Open
Abstract
Neurogenin 1 (Ngn1) belongs to the basic helix–loop–helix (bHLH) transcription factor family and plays important roles in specifying neuronal differentiation. The present study aimed to determine whether forced Ngn1 expression contributes to bone homeostasis. Ngn1 inhibited the p300/CREB-binding protein-associated factor (PCAF)-induced acetylation of nuclear factor of activated T cells 1 (NFATc1) and runt-related transcription factor 2 (Runx2) through binding to PCAF, which led to the inhibition of osteoclast and osteoblast differentiation, respectively. In addition, Ngn1 overexpression inhibited the TNF-α- and IL-17A-mediated enhancement of osteoclast differentiation and IL-17A-induced osteoblast differentiation. These findings indicate that Ngn1 can serve as a novel therapeutic agent for treating ankylosing spondylitis with abnormally increased bone formation and resorption.
Collapse
Affiliation(s)
- Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
| | - Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
| | - Jeong-Tae Koh
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
- Correspondence: ; Tel.: +82-61-379-2835
| |
Collapse
|
30
|
Qin Q, Gomez-Salazar M, Cherief M, Pagani CA, Lee S, Hwang C, Tower RJ, Onggo S, Sun Y, Piplani A, Li Z, Ramesh S, Clemens TL, Levi B, James AW. Neuron-to-vessel signaling is a required feature of aberrant stem cell commitment after soft tissue trauma. Bone Res 2022; 10:43. [PMID: 35641477 PMCID: PMC9156761 DOI: 10.1038/s41413-022-00216-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/04/2022] [Accepted: 04/29/2022] [Indexed: 11/10/2022] Open
Abstract
The functional interdependence of nerves and blood vessels is a well-established concept during tissue morphogenesis, yet the role of neurovascular coupling in proper and aberrant tissue repair is an emerging field of interest. Here, we sought to define the regulatory relationship of peripheral nerves on vasculature in a severe extremity trauma model in mice, which results in aberrant cell fate and heterotopic ossification (HO). First, a high spatial degree of neurovascular congruency was observed to exist within extremity injury associated heterotopic ossification. Vascular and perivascular cells demonstrate characteristic responses to injury, as assessed by single cell RNA sequencing. This vascular response to injury was blunted in neurectomized mice, including a decrease in endothelial proliferation and type H vessel formation, and a downregulation of key transcriptional networks associated with angiogenesis. Independent mechanisms to chemically or genetically inhibit axonal ingrowth led to similar deficits in HO site angiogenesis, a reduction in type H vessels, and heterotopic bone formation. Finally, a combination of single cell transcriptomic approaches within the dorsal root ganglia identified key neural-derived angiogenic paracrine factors that may mediate neuron-to-vascular signaling in HO. These data provide further understanding of nerve-to-vessel crosstalk in traumatized soft tissues, which may reflect a key determinant of mesenchymal progenitor cell fate after injury.
Collapse
Affiliation(s)
- Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mario Gomez-Salazar
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Chase A Pagani
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas, Southwestern, TX, USA
| | - Seungyong Lee
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Charles Hwang
- Department of Plastic Surgery, Harvard, Cambridge, MA, 02138, USA
| | - Robert J Tower
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas, Southwestern, TX, USA
- Department of Orthopaedics, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Sharon Onggo
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yuxiao Sun
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas, Southwestern, TX, USA
| | - Abhinav Piplani
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Sowmya Ramesh
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Thomas L Clemens
- Department of Orthopaedics, Johns Hopkins University, Baltimore, MD, 21205, USA
- Baltimore Veterans Administration Medical Center, Baltimore, MD, 21201, USA
| | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas, Southwestern, TX, USA.
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
31
|
Nakanishi Y, Kang S, Kumanogoh A. Crosstalk between axon guidance signaling and bone remodeling. Bone 2022; 157:116305. [PMID: 34973495 DOI: 10.1016/j.bone.2021.116305] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 01/04/2023]
Abstract
The maintenance of skeletal integrity is tightly regulated by two cell types, bone forming osteoblasts and bone resorbing osteoclasts. Although the role of the nervous system in regulating osteoblasts and osteoclasts was identified over a decade ago, the molecular mechanism of skeletal-neural interactions in bone homeostasis has only been studied recently. In particular, the complex roles of axon guidance molecules, such as semaphorins and ephrins, in the bone have been studied extensively. In this review, we highlight the latest advances in determining the functions of semaphorins and ephrins in the establishment and maintenance of the skeletal system, with a focus on the functional interaction between the skeletal and nervous systems.
Collapse
Affiliation(s)
- Yoshimitsu Nakanishi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita City, Osaka 565-0871, Japan
| | - Sujin Kang
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita City, Osaka 565-0871, Japan; Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita City, Osaka 565-0871, Japan; Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
32
|
Yan Z, Li K, Shao D, Shen Q, Ding Y, Huang S, Xie Y, Zheng X. Visible-light-responsive reduced graphene oxide/g-C 3N 4/TiO 2 composite nanocoating for photoelectric stimulation of neuronal and osteoblastic differentiation. RSC Adv 2022; 12:8878-8888. [PMID: 35424887 PMCID: PMC8985170 DOI: 10.1039/d2ra00282e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/08/2022] [Indexed: 11/21/2022] Open
Abstract
rGO/g-C3N4/TiO2 nanocoating was fabricated on Ti-based implant for photoelectric stimulation of bone and nerve repair. The ternary nanocoating exerted greater photoelectric effects on enhancing osteoblastic differentiation and neurite outgrowth.
Collapse
Affiliation(s)
- Ziru Yan
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Kai Li
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Dandan Shao
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Qingyi Shen
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi Ding
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Shansong Huang
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Youtao Xie
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Xuebin Zheng
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
33
|
Jiang Y, Xin N, Xiong Y, Guo Y, Yuan Y, Zhang Q, Gong P. αCGRP Regulates Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells Through ERK1/2 and p38 MAPK Signaling Pathways. Cell Transplant 2022; 31:9636897221107636. [PMID: 35758252 PMCID: PMC9247368 DOI: 10.1177/09636897221107636] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
As a typical neuropeptide richly distributed in central and peripheral nervous
systems, α-calcitonin-gene-related peptide (αCGRP) has recently been found to
play a crucial role in bone development and metabolism, but the mechanisms
involved are not fully uncovered. Here, this study aimed to investigate the
effects and underlying molecular mechanisms of αCGRP in regulating the
osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Using
microarray technology, gene ontology (GO) and kyoto encyclopedia of genes and
genomes (KEGG) analyses revealed that osteogenic properties of BMSCs were
facilitated and mitogen-activated protein kinase (MAPK) signaling pathway was
upregulated by αCGRP in this process. Through western blot assay, we proved that
αCGRP led to an increased phosphorylation level of extracellular
signal-regulated kinases 1 and 2 (ERK1/2) and p38 MAPK signaling cascades in a
time-dependent manner. And αCGRP could promote differentiative capacity of
BMSCs, showing upregulated mRNA and protein expression level of alkaline
phosphatase (Alp), collagen type 1 (Col-1), osteopontin (Opn), and runt-related
transcription factor 2 (Runx2), as well as increased ALP activity and calcified
nodules. The addition of ERK1/2 or p38 MAPK inhibitor—U0126 or SB203580,
resulted in an impaired osteogenic differentiation of BMSCs. Besides,
inactivation of this signal transduction had negative impacts on proliferative
activity and apoptotic process of αCGRP-mediated BMSCs. Our findings
demonstrated that MAPK signaling pathway, at least in part, was responsible for
the enhanced BMSCs’ osteogenesis induced by αCGRP, which might offer us
promising strategies for bone-related disorders.
Collapse
Affiliation(s)
- Yixuan Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Na Xin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanjun Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Jinjiang Out-Patient Section, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ying Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Qi J, Yu T, Hu B, Wu H, Ouyang H. Current Biomaterial-Based Bone Tissue Engineering and Translational Medicine. Int J Mol Sci 2021; 22:10233. [PMID: 34638571 PMCID: PMC8508818 DOI: 10.3390/ijms221910233] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/14/2021] [Accepted: 09/19/2021] [Indexed: 11/16/2022] Open
Abstract
Bone defects cause significant socio-economic costs worldwide, while the clinical "gold standard" of bone repair, the autologous bone graft, has limitations including limited graft supply, secondary injury, chronic pain and infection. Therefore, to reduce surgical complexity and speed up bone healing, innovative therapies are needed. Bone tissue engineering (BTE), a new cross-disciplinary science arisen in the 21st century, creates artificial environments specially constructed to facilitate bone regeneration and growth. By combining stem cells, scaffolds and growth factors, BTE fabricates biological substitutes to restore the functions of injured bone. Although BTE has made many valuable achievements, there remain some unsolved challenges. In this review, the latest research and application of stem cells, scaffolds, and growth factors in BTE are summarized with the aim of providing references for the clinical application of BTE.
Collapse
Affiliation(s)
- Jingqi Qi
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang University-University of Edinburgh Institute, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Tianqi Yu
- Department of Mechanical Engineering, Zhejiang University-University of Illinois at Urbana-Champaign Institute, Zhejiang University, Haining 314400, China;
| | - Bangyan Hu
- Section of Molecular and Cell Biology, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA;
| | - Hongwei Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang University-University of Edinburgh Institute, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang University-University of Edinburgh Institute, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310003, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou 310003, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310003, China
| |
Collapse
|
35
|
Imbrici P, Accogli A, Blunck R, Altamura C, Iacomino M, D’Adamo MC, Allegri A, Pedemonte M, Brolatti N, Vari S, Cataldi M, Capra V, Gustincich S, Zara F, Desaphy JF, Fiorillo C. Musculoskeletal Features without Ataxia Associated with a Novel de novo Mutation in KCNA1 Impairing the Voltage Sensitivity of Kv1.1 Channel. Biomedicines 2021; 9:75. [PMID: 33466780 PMCID: PMC7829709 DOI: 10.3390/biomedicines9010075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
The KCNA1 gene encodes the α subunit of the voltage-gated Kv1.1 potassium channel that critically regulates neuronal excitability in the central and peripheral nervous systems. Mutations in KCNA1 have been classically associated with episodic ataxia type 1 (EA1), a movement disorder triggered by physical and emotional stress. Additional features variably reported in recent years include epilepsy, myokymia, migraine, paroxysmal dyskinesia, hyperthermia, hypomagnesemia, and cataplexy. Interestingly, a few individuals with neuromyotonia, either isolated or associated with skeletal deformities, have been reported carrying variants in the S2-S3 transmembrane segments of Kv1.1 channels in the absence of any other symptoms. Here, we have identified by whole-exome sequencing a novel de novo variant, T268K, in KCNA1 in a boy displaying recurrent episodes of neuromyotonia, muscle hypertrophy, and skeletal deformities. Through functional analysis in heterologous cells and structural modeling, we show that the mutation, located at the extracellular end of the S3 helix, causes deleterious effects, disrupting Kv1.1 function by altering the voltage dependence of activation and kinetics of deactivation, likely due to abnormal interactions with the voltage sensor in the S4 segment. Our study supports previous evidence suggesting that specific residues within the S2 and S3 segments of Kv1.1 result in a distinctive phenotype with predominant musculoskeletal presentation.
Collapse
Affiliation(s)
- Paola Imbrici
- Department of Pharmacy-Drug Sciences, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Andrea Accogli
- Medical Genetics Unit, IRCCS Institute “G. Gaslini”, 80131 Genoa, Italy; (A.A.); (M.I.)
| | - Rikard Blunck
- Department of Physics, Université de Montréal, Montréal, QC H3C 3J7, Canada;
| | - Concetta Altamura
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70121 Bari, Italy; (C.A.); (J.-F.D.)
| | - Michele Iacomino
- Medical Genetics Unit, IRCCS Institute “G. Gaslini”, 80131 Genoa, Italy; (A.A.); (M.I.)
| | - Maria Cristina D’Adamo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MDS-2080 Msida, Malta;
| | - Anna Allegri
- Paediatric Endocrinology Unit, IRCCS Institute “G. Gaslini”, 80131 Genoa, Italy;
| | - Marina Pedemonte
- Paediatric Neurology and Neuromuscular Disorders Unit, IRCCS Institute “G. Gaslini”, 80131 Genoa, Italy; (M.P.); (N.B.); (S.V.); (C.F.)
| | - Noemi Brolatti
- Paediatric Neurology and Neuromuscular Disorders Unit, IRCCS Institute “G. Gaslini”, 80131 Genoa, Italy; (M.P.); (N.B.); (S.V.); (C.F.)
| | - Stella Vari
- Paediatric Neurology and Neuromuscular Disorders Unit, IRCCS Institute “G. Gaslini”, 80131 Genoa, Italy; (M.P.); (N.B.); (S.V.); (C.F.)
| | - Matteo Cataldi
- Neuropsychiatric Unit, IRCCS Institute “G. Gaslini”, 80131 Genoa, Italy;
| | - Valeria Capra
- Neurosurgery Unit, IRCCS Institute “G. Gaslini”, 80131 Genoa, Italy;
| | - Stefano Gustincich
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 80131 Genoa, Italy;
| | - Federico Zara
- Medical Genetics Unit, IRCCS Institute “G. Gaslini”, 80131 Genoa, Italy; (A.A.); (M.I.)
| | - Jean-Francois Desaphy
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70121 Bari, Italy; (C.A.); (J.-F.D.)
| | - Chiara Fiorillo
- Paediatric Neurology and Neuromuscular Disorders Unit, IRCCS Institute “G. Gaslini”, 80131 Genoa, Italy; (M.P.); (N.B.); (S.V.); (C.F.)
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
| |
Collapse
|
36
|
Cartilage Targets of Knee Osteoarthritis Shared by Both Genders. Int J Mol Sci 2021; 22:ijms22020569. [PMID: 33430025 PMCID: PMC7827374 DOI: 10.3390/ijms22020569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022] Open
Abstract
As the leading cause of disability, osteoarthritis (OA) affects people of all ages, sexes, and races. With the increasing understanding of OA, the sex differences have attracted specific attention as the burden of OA is greater in women. There is no doubt that gender-specific OA management has great potential for precision treatment. On the other hand, from the marketing aspect, a medication targeting the OA-responsive biomarker(s) shared by both genders is more favorable for drug development. Thus, in the current study, a published transcriptome dataset of knee articular cartilage was used to compare OA and healthy samples for identifying the genes with the same significantly different expression trend in both males and females. With 128 genes upregulated and 143 genes downregulated in both OA males and females, 9 KEGG pathways have been enriched based on the current knowledge, including 'renal cell carcinoma,' 'ECM-receptor interaction,' 'HIF-1 signaling pathway,' 'MicroRNAs in cancer,' 'focal adhesion,' 'Relaxin signaling pathway,' 'breast cancer,' 'PI3K-Akt signaling pathway,' and 'human papillomavirus infection.' Here, we explore the potential impacts of these clusters in OA. We also analyze the identified 'cell plasma membrane related genes' in-depth to identify the potential chondrocyte cell surface target(s) of OA management.
Collapse
|
37
|
Guo X, Feng Y, Sun T, Feng S, Tang J, Chen L, Cao X, Lin H, He X, Li M, Zhang Z, Yin G, Mei X, Huang H. Clinical guidelines for neurorestorative therapies in spinal cord injury (2021 China version). JOURNAL OF NEURORESTORATOLOGY 2021. [DOI: 10.26599/jnr.2021.9040003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Treatment of spinal cord injury (SCI) remains challenging. Considering the rapid developments in neurorestorative therapies for SCI, we have revised and updated the Clinical Therapeutic Guidelines for Neurorestoration in Spinal Cord Injury (2016 Chinese version) of the Chinese Association of Neurorestoratology (Preparatory) and China Committee of International Association of Neurorestoratology. Treatment of SCI is a systematic multimodal process that aims to improve survival and restore neurological function. These guidelines cover real-world comprehensive neurorestorative management of acute, subacute, and chronic SCI and include assessment and diagnosis, pre-hospital first aid, treatment, rehabilitation, and complication management.
Collapse
|
38
|
Arthur A, Gronthos S. Clinical Application of Bone Marrow Mesenchymal Stem/Stromal Cells to Repair Skeletal Tissue. Int J Mol Sci 2020; 21:E9759. [PMID: 33371306 PMCID: PMC7767389 DOI: 10.3390/ijms21249759] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
There has been an escalation in reports over the last decade examining the efficacy of bone marrow derived mesenchymal stem/stromal cells (BMSC) in bone tissue engineering and regenerative medicine-based applications. The multipotent differentiation potential, myelosupportive capacity, anti-inflammatory and immune-modulatory properties of BMSC underpins their versatile nature as therapeutic agents. This review addresses the current limitations and challenges of exogenous autologous and allogeneic BMSC based regenerative skeletal therapies in combination with bioactive molecules, cellular derivatives, genetic manipulation, biocompatible hydrogels, solid and composite scaffolds. The review highlights the current approaches and recent developments in utilizing endogenous BMSC activation or exogenous BMSC for the repair of long bone and vertebrae fractures due to osteoporosis or trauma. Current advances employing BMSC based therapies for bone regeneration of craniofacial defects is also discussed. Moreover, this review discusses the latest developments utilizing BMSC therapies in the preclinical and clinical settings, including the treatment of bone related diseases such as Osteogenesis Imperfecta.
Collapse
Affiliation(s)
- Agnieszka Arthur
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5001, Australia;
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5001, Australia;
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| |
Collapse
|