1
|
Ruan X, Jin X, Sun F, Pi J, Jinghu Y, Lin X, Zhang N, Chen G. IGF signaling pathway in bone and cartilage development, homeostasis, and disease. FASEB J 2024; 38:e70031. [PMID: 39206513 DOI: 10.1096/fj.202401298r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The skeleton plays a fundamental role in the maintenance of organ function and daily activities. The insulin-like growth factor (IGF) family is a group of polypeptide substances with a pronounced role in osteoblast differentiation, bone development, and metabolism. Disturbance of the IGFs and the IGF signaling pathway is inextricably linked with assorted developmental defects, growth irregularities, and jeopardized skeletal structure. Recent findings have illustrated the significance of the action of the IGF signaling pathway via growth factors and receptors and its interactions with dissimilar signaling pathways (Wnt/β-catenin, BMP, TGF-β, and Hh/PTH signaling pathways) in promoting the growth, survival, and differentiation of osteoblasts. IGF signaling also exhibits profound influences on cartilage and bone development and skeletal homeostasis via versatile cell-cell interactions in an autocrine, paracrine, and endocrine manner systemically and locally. Our review summarizes the role and regulatory function as well as a potentially integrated gene network of the IGF signaling pathway with other signaling pathways in bone and cartilage development and skeletal homeostasis, which in turn provides an enlightening insight into visualizing bright molecular targets to be eligible for designing effective drugs to handle bone diseases and maladies, such as osteoporosis, osteoarthritis, and dwarfism.
Collapse
Affiliation(s)
- Xinyi Ruan
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiuhui Jin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Fuju Sun
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiashun Pi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yihan Jinghu
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xinyi Lin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nenghua Zhang
- Clinical Laboratory, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
2
|
Xing W, Kesavan C, Pourteymoor S, Mohan S. Global and Conditional Disruption of the Igf-I Gene in Osteoblasts and/or Chondrocytes Unveils Epiphyseal and Metaphyseal Bone-Specific Effects of IGF-I in Bone. BIOLOGY 2023; 12:1228. [PMID: 37759627 PMCID: PMC10525837 DOI: 10.3390/biology12091228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/24/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
To evaluate the relative importance of IGF-I expression in various cell types for endochondral ossification, we quantified the trabecular bone at the secondary spongiosa and epiphysis of the distal femur in 8-12-week-old male mice with a global knockout of the Igf-I gene, as well as the conditional deletion of Igf-I in osteoblasts, chondrocytes, and osteoblasts/chondrocytes and their corresponding wild-type control littermates. The osteoblast-, chondrocyte-, and osteoblast/chondrocyte-specific Igf-I conditional knockout mice were generated by crossing Igf-I floxed mice with Cre transgenic mice in which Cre expression is under the control of either the Col1α2 or Col2α1 promoter. We found that the global disruption of Igf-I resulted in 80% and 70% reductions in bone size, defined as total volume, at the secondary spongiosa and epiphysis of the distal femur, respectively. The abrogation of Igf-I in Col1α2-producing osteoblasts but not Col2α1-producing chondrocytes decreased bone size by 25% at both the secondary spongiosa and epiphysis. In comparison, the deletion of the Igf-I globally or specifically in osteoblasts or chondrocytes reduced trabecular bone mass by 25%. In contrast, the universal deletion of Igf-I in all cells, but not the conditional disruption of Igf-I in osteoblasts and/or chondrocytes reduced trabecular bone mass in the epiphysis. The reduced trabecular bone mass at the secondary spongiosa in osteoblast- and/or chondrocyte-specific Igf-I conditional knockout mice is caused by the reduced trabecular number and increased trabecular separation. Immunohistochemistry studies found that the expression levels of chondrocyte (COL10, MMP13) and osteoblast (BSP) markers were less in the secondary spongiosa and the epiphyses in the global Igf-I deletion mice. Our data indicate that local and endocrine Igf-I act pleiotropically and in a cell type- and bone compartment-dependent manner in bone.
Collapse
Affiliation(s)
- Weirong Xing
- VA Loma Linda Healthcare Systems, Musculoskeletal Disease Center, Loma Linda, CA 92357, USA; (W.X.); (C.K.); (S.P.)
- Departments of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Chandrasekhar Kesavan
- VA Loma Linda Healthcare Systems, Musculoskeletal Disease Center, Loma Linda, CA 92357, USA; (W.X.); (C.K.); (S.P.)
- Departments of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Sheila Pourteymoor
- VA Loma Linda Healthcare Systems, Musculoskeletal Disease Center, Loma Linda, CA 92357, USA; (W.X.); (C.K.); (S.P.)
| | - Subburaman Mohan
- VA Loma Linda Healthcare Systems, Musculoskeletal Disease Center, Loma Linda, CA 92357, USA; (W.X.); (C.K.); (S.P.)
- Departments of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Departments of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA
- Departments of Orthopedic Surgery, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
3
|
Abstract
Although osteoblasts and osteocytes are descended from the same lineage, they each have unique and essential roles in bone. Targeting gene deletion to osteoblasts and osteocytes using the Cre/loxP system has greatly increased our current understanding of how these cells function. Additionally, the use of the Cre/loxP system in conjunction with cell-specific reporters has enabled lineage tracing of these bone cells both in vivo and ex vivo. However, concerns have been raised regarding the specificity of the promoters used and the resulting off-target effects on cells within and outside of the bone. In this review, we have summarized the main mouse models that have been used to determine the functions of specific genes in osteoblasts and osteocytes. We discuss the expression patterns and specificity of the different promoter fragments during osteoblast to osteocyte differentiation in vivo. We also highlight how their expression in non-skeletal tissues may complicate the interpretation of study results. A thorough understanding of when and where these promoters are activated will enable improved study design and greater confidence in data interpretation.
Collapse
Affiliation(s)
- Y Kitase
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States of America
| | - M Prideaux
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States of America.
| |
Collapse
|
4
|
Esposito A, Klüppel M, Wilson BM, Meka SRK, Spagnoli A. CXCR4 mediates the effects of IGF-1R signaling in rodent bone homeostasis and fracture repair. Bone 2023; 166:116600. [PMID: 36368465 PMCID: PMC10057209 DOI: 10.1016/j.bone.2022.116600] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/10/2022]
Abstract
Non-union fractures have considerable clinical and economic burdens and yet the underlying pathogenesis remains largely undetermined. The fracture healing process involves cellular differentiation, callus formation and remodeling, and implies the recruitment and differentiation of mesenchymal stem cells that are not fully characterized. C-X-C chemokine receptor 4 (CXCR4) and Insulin-like growth factor 1 receptor (IGF-1R) are expressed in the fracture callus, but their interactions still remain elusive. We hypothesized that the regulation of CXCR4 by IGF-1R signaling is essential to maintain the bone homeostasis and to promote fracture repair. By using a combination of in vivo and in vitro approaches, we found that conditional ablation of IGF-1R in osteochondroprogenitors led to defects in bone formation and mineralization that associated with altered expression of CXCR4 by a discrete population of endosteal cells. These defects were corrected by AMD3100 (a CXCR4 antagonist). Furthermore, we found that the inducible ablation of IGF-1R in osteochondroprogenitors led to fracture healing failure, that associated with an altered expression of CXCR4. In vivo AMD3100 treatment improved fracture healing and normalized CXCR4 expression. Moreover, we determined that these effects were mediated through the IGF-1R/Insulin receptor substrate 1 (IRS-1) signaling pathway. Taken together, our studies identified a novel population of endosteal cells that is functionally regulated through the modulation of CXCR4 by IGF-1R signaling, and such control is essential in bone homeostasis and fracture healing. Knowledge gained from these studies has the potential to accelerate the development of novel therapeutic interventions by targeting CXCR4 signaling to treat non-unions.
Collapse
Affiliation(s)
- Alessandra Esposito
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Michael Klüppel
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Brittany M Wilson
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Sai R K Meka
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Anna Spagnoli
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA; Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
5
|
Casanova M, Schindeler A, Peacock L, Lee L, Schneider P, Little DG, Müller R. Characterization of the Developing Lacunocanalicular Network During Fracture Repair. JBMR Plus 2021; 5:e10525. [PMID: 34532613 PMCID: PMC8441443 DOI: 10.1002/jbm4.10525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/23/2021] [Accepted: 06/06/2021] [Indexed: 11/09/2022] Open
Abstract
Fracture repair is a normal physiological response to bone injury. During the process of bony callus formation, a lacunocanalicular network (LCN) is formed de novo that evolves with callus remodeling. Our aim was the longitudinal assessment of the development and evolution of the LCN during fracture repair. To this end, 45 adult wild‐type C57BL/6 mice underwent closed tibial fracture surgery. Fractured and intact contralateral tibias were harvested after 2, 3, and 6 weeks of bone healing (n = 15/group). High‐resolution micro–computed tomography (μCT) and deconvolution microscopy (DV) approaches were applied to quantify lacunar number density from the calluses and intact bone. On histological sections, Goldner's trichrome staining was used to assess lacunar occupancy, fluorescein isothiocyanate staining to visualize the canalicular network, and terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate‐biotin nick end labeling (TUNEL) staining to examine osteocyte apoptosis. Analysis of μCT scans showed progressive decreases in mean lacuna volume over time (−27% 2–3 weeks; −13% 3–6 weeks). Lacunar number density increased considerably between 2 and 3 weeks (+156%). Correlation analysis was performed, showing a positive linear relationship between canalicular number density and trabecular thickness (R2 = 0.56, p < 0.001) and an inverse relationship between mean lacuna volume and trabecular thickness (R2 = 0.57, p < 0.001). Histology showed increases in canalicular number density over time (+22% 2–3 weeks, +51% 3–6 weeks). Lacunar occupancy in new bone of the callus was high (>90%), but the old cortical bone within the fracture site appeared necrotic as it underwent resorption. In conclusion, our data shows a progressive increase in the complexity of the LCN over time during fracture healing and demonstrates that this network is initiated during the early stages of repair. Further studies are needed to address the functional importance of osteocytes in bone healing, particularly in detecting and translating the effects of micromotion in the fracture. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology The Children's Hospital at Westmead Westmead Australia.,Discipline of Child and Adolescent Health University of Sydney Camperdown Australia
| | - Lauren Peacock
- Orthopaedic Research & Biotechnology The Children's Hospital at Westmead Westmead Australia
| | - Lucinda Lee
- Orthopaedic Research & Biotechnology The Children's Hospital at Westmead Westmead Australia.,Discipline of Child and Adolescent Health University of Sydney Camperdown Australia
| | - Philipp Schneider
- Institute for Biomechanics ETH Zurich Zurich Switzerland.,Bioengineering Science Research Group, Faculty of Engineering and Physical Sciences University of Southampton Southampton UK.,High-Performance Vision Systems, Center for Vision, Automation & Control Austrian Institute of Technology (AIT) Vienna Austria
| | - David G Little
- Orthopaedic Research & Biotechnology The Children's Hospital at Westmead Westmead Australia.,Discipline of Child and Adolescent Health University of Sydney Camperdown Australia
| | - Ralph Müller
- Institute for Biomechanics ETH Zurich Zurich Switzerland
| |
Collapse
|
6
|
Tucci MA, Pride Y, Strickland S, Marocho SMS, Jackson RJ, Jefferson JR, Chade AR, Grill RJ, Grayson BE. Delayed Systemic Treatment with Cannabinoid Receptor 2 Agonist Mitigates Spinal Cord Injury-Induced Osteoporosis More Than Acute Treatment Directly after Injury. Neurotrauma Rep 2021; 2:270-284. [PMID: 34223557 PMCID: PMC8244511 DOI: 10.1089/neur.2020.0059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Nearly all persons with spinal cord injury (SCI) will develop osteoporosis following injury, and further, up to 50% of all persons with SCI will sustain a fracture during their lives. The unique mechanisms driving osteoporosis following SCI remain unknown. The cannabinoid system modulation of bone metabolism through cannabinoid 1/2 (CB1/2) has been of increasing interest for the preservation of bone mass and density in models of osteoporosis. Using a thoracic vertebral level 8 (T8) complete transection in a mouse model, we performed daily treatment with a selective CB2 receptor agonist, HU308, compared with SCI-vehicle-treated and naïve control animals either immediately after injury for 40 days, or in a delayed paradigm, following 3 months after injury. The goal was to prevent or potentially reverse SCI-induced osteoporosis. In the acute phase, administration of the CB2 agonist was not able to preserve the rapid loss of cancellous bone. In the delayed-treatment paradigm, in cortical bone, HU308 increased cortical-area to total-area ratio and periosteal perimeter in the femur, and improved bone density in the distal femur and proximal tibia. Further, we report changes to the metaphyseal periosteum with increased presence of adipocyte and fat mass in the periosteum of SCI animals, which was not present in naïve animals. The layer of fat increased markedly in HU308-treated animals compared with SCI-vehicle-treated animals. Overall, these data show that CB2 agonism targets a number of cell types that can influence overall bone quality.
Collapse
Affiliation(s)
- Michelle A. Tucci
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Yilianys Pride
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Suzanne Strickland
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Susanna M. Salazar Marocho
- Department of Biomedical Materials Science, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ramon J. Jackson
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Joshua R. Jefferson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Alejandro R. Chade
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Radiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Raymond J. Grill
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Bernadette E. Grayson
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
7
|
Comeau-Gauthier M, Tarchala M, Luna JLRG, Harvey E, Merle G. Unleashing β-catenin with a new anti-Alzheimer drug for bone tissue regeneration. Injury 2020; 51:2449-2459. [PMID: 32829895 DOI: 10.1016/j.injury.2020.07.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/18/2020] [Indexed: 02/02/2023]
Abstract
The Wnt/β-catenin signaling pathway is critical for bone differentiation and regeneration. Tideglusib, a selective FDA approved glycogen synthase kinase-3β (GSK-3β) inhibitor, has been shown to promote dentine formation, but its effect on bone has not been examined. Our objective was to study the effect of localized Tideglusib administration on bone repair. Bone healing between Tideglusib treated and control mice was analysed at 7, 14 and 28 days postoperative (PO) with microCT, dynamic histomorphometry and immunohistology. There was a local downregulation of GSK-3β in Tideglusib animals, resulting in a significant increase in the amount of new bone formation with both enhanced cortical bone bridging and medullary bone deposition. The bone formation in the Tideglusib group was characterized by early osteoblast differentiation with down-regulation of GSK-3β at day 7 and 14, and higher accumulation of active β-catenin at day 14. Here, for the first time, we show a positive effect of Tideglusib on bone formation through the inactivation of GSK-3β. Furthermore, the findings suggest that Tideglusib does not interfere with precursor cell recruitment and commitment, contrary to other GSK-3β antagonists such as lithium chloride. Taken together, the results indicate that Tideglusib could be used directly at a fracture site during the initial intraoperative internal fixation without the need for further surgery, injection or drug delivery system. This FDA-approved drug may be useful in the future for the prevention of non-union in patients presenting with a high risk for fracture-healing.
Collapse
Affiliation(s)
- Marianne Comeau-Gauthier
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal, Canada; Experimental Surgery, Faculty of Medicine, McGill University. Rue de la Montaigne, Montreal, QC, Canada.
| | - Magdalena Tarchala
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal, Canada; Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Qc., H3G 1A4 Canada.
| | - Jose Luis Ramirez-Garcia Luna
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal, Canada; Experimental Surgery, Faculty of Medicine, McGill University. Rue de la Montaigne, Montreal, QC, Canada; Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Qc., H3G 1A4 Canada.
| | - Edward Harvey
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal, Canada; Bone Engineering Labs, Montreal General Hospital, 1650 Cedar Avenue, Room C10-124, Montreal, Qc., H3G 1A4 Canada.
| | - Geraldine Merle
- Chemical Engineering Department, Polytechnique J.-A.-Bombardier building Polytechnique Montréal C.P. 6079, succ. Centre-ville, Montréal (Québec), H3C 3A7, Canada.
| |
Collapse
|
8
|
Kerschan-Schindl K, Tiefenböck TM, Föger-Samwald U, Payr S, Frenzel S, Marculescu R, Gleiss A, Sarahrudi K, Pietschmann P. Circulating Myostatin Levels Decrease Transiently after Implantation of a Hip Hemi-Arthroplasty. Gerontology 2020; 66:393-400. [PMID: 32454508 DOI: 10.1159/000507731] [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: 11/13/2019] [Accepted: 04/03/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Muscle and bone metabolism are both important for the healing of fractures and the regeneration of injured muscle tissue. The aim of this investigation was to evaluate myostatin and other regulating factors in patients with hip fractures who underwent hemi-arthroplasty. METHODS Serum levels of myostatin (MSTN), follistatin (FSTN), dickkopf-1 (Dkk1), and periostin (PSTN) as well as markers of bone turnover were evaluated in patients with hip fractures before surgery and twice in the 2 weeks after surgery. These parameters were also evaluated in age- and gender-matched subjects without major musculoskeletal injury. RESULTS MSTN was transiently reduced; its opponent FSTN was transiently increased. Dkk1, the negative regulator of bone mass, and PSTN, a marker of subperiosteal bone formation, increased after surgery. With regard to markers of bone turnover, resorption was elevated during the entire period of observation whereas the early bone formation marker N-terminal propeptide of type I collagen was elevated 12 days after surgery. CONCLUSIONS Unexpectedly, MSTN, a negative regulator of muscle growth, was reduced after surgery compared with before surgery. As musculoskeletal markers are altered during bone healing, they do not reflect general bone metabolism after fracture or joint arthroplasty. This is important because many elderly patients receive treatment for osteoporosis.
Collapse
Affiliation(s)
- Katharina Kerschan-Schindl
- Department of Physical Medicine, Rehabilitation and Occupational Therapy, Medical University of Vienna, Vienna, Austria
| | - Thomas M Tiefenböck
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Ursula Föger-Samwald
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Stephan Payr
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Stephan Frenzel
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Andreas Gleiss
- Center of Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Kambiz Sarahrudi
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria, .,Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria,
| | - Peter Pietschmann
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Sun X, Gao X, Deng Z, Zhang L, McGilvray K, Gadomski BC, Amra S, Bao G, Huard J. High bone microarchitecture, strength, and resistance to bone loss in MRL/MpJ mice correlates with activation of different signaling pathways and systemic factors. FASEB J 2019; 34:789-806. [PMID: 31914651 DOI: 10.1096/fj.201901229rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/30/2019] [Accepted: 11/02/2019] [Indexed: 12/27/2022]
Abstract
The MRL/MpJ mice have demonstrated an enhanced tissue regeneration capacity for various tissues. In the present study, we systematically characterized bone microarchitecture and found that MRL/MpJ mice exhibit higher bone microarchitecture and strength compared to both C57BL/10J and C57BL/6J WT mice at 2, 4, and 10 months of age. The higher bone mass in MRL/MpJ mice was correlated to increased osteoblasts, decreased osteoclasts, higher cell proliferation, and bone formation, and enhanced pSMAD5 signaling earlier during postnatal development (2-month old) in the spine trabecular bone, and lower bone resorption rate at later age. Furthermore, these mice exhibit accelerated fracture healing via enhanced pSMAD5, pAKT and p-P38MAPK pathways compared to control groups. Moreover, MRL/MpJ mice demonstrated resistance to ovariectomy-induced bone loss as evidenced by maintaining higher bone volume/tissue volume (BV/TV) and lower percentage of bone loss later after ovariectomy. The consistently higher serum IGF1 level and lower RANKL level in MRL/MpJ mice may contribute to the maintenance of high bone mass in uninjured and injured bone. In conclusion, our results indicate that enhanced pSMAD5, pAKT, and p-P38MAPK signaling, higher serum IGF-1, and lower RANKL level contribute to the higher bone microarchitecture and strength, accelerated healing, and resistance to osteoporosis in MRL/MpJ mice.
Collapse
Affiliation(s)
- Xuying Sun
- Department of Orthopaedic Surgery, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Xueqin Gao
- Department of Orthopaedic Surgery, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado
| | - Zhenhan Deng
- Department of Orthopaedic Surgery, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Linlin Zhang
- Department of Biomedical Engineering, Rice University, Houston, Texas
| | - Kirk McGilvray
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Benjamin C Gadomski
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Sarah Amra
- Department of Orthopaedic Surgery, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Gang Bao
- Department of Biomedical Engineering, Rice University, Houston, Texas
| | - Johnny Huard
- Department of Orthopaedic Surgery, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado
| |
Collapse
|
10
|
Choy MHV, Wong RMY, Chow SKH, Li MC, Chim YN, Li TK, Ho WT, Cheng JCY, Cheung WH. How much do we know about the role of osteocytes in different phases of fracture healing? A systematic review. J Orthop Translat 2019; 21:111-121. [PMID: 32309136 PMCID: PMC7152791 DOI: 10.1016/j.jot.2019.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/22/2019] [Accepted: 07/15/2019] [Indexed: 12/21/2022] Open
Abstract
Background Although emerging studies have provided evidence that osteocytes are actively involved in fracture healing, there is a general lack of a detailed understanding of the mechanistic pathway, cellular events and expression of markers at different phases of healing. Methods This systematic review describes the role of osteocytes in fracture healing from early to late phase. Literature search was performed in PubMed and Embase. Original animal and clinical studies with available English full-text were included. Information was retrieved from the selected studies. Results A total of 23 articles were selected in this systematic review. Most of the studies investigated changes of various genes and proteins expression patterns related to osteocytes. Several studies have described a constant expression of osteocyte-specific marker genes throughout the fracture healing cascade followed by decline phase with the progress of healing, denoting the important physiological role of the osteocyte and the osteocyte lacuno-canalicular network in fracture healing. The reports of various markers suggested that osteocytes could trigger coordinated bone healing responses from cell death and expression of proinflammatory markers cyclooxygenase-2 and interleukin 6 at early phase of fracture healing. This is followed by the expression of growth factors bone morphogenetic protein-2 and cysteine-rich angiogenic inducer 61 that matched with the neo-angiogenesis, chondrogenesis and callus formation during the intermediate phase. Tightly controlled regulation of osteocyte-specific markers E11/Podoplanin (E11), dentin matrix protein 1 and sclerostin modulate and promote osteogenesis, mineralisation and remodelling across different phases of fracture healing. Stabilised fixation was associated with the finding of higher number of osteocytes with little detectable bone morphogenetic proteins expressions in osteocytes. Sclerostin-antibody treatment was found to result in improvement in bone mass, bone strength and mineralisation. Conclusion To further illustrate the function of osteocytes, additional longitudinal studies with appropriate clinically relevant model to study osteoporotic fractures are crucial. Future investigations on the morphological changes of osteocyte lacuno-canalicular network during healing, osteocyte-mediated signalling molecules in the transforming growth factor-beta-Smad3 pathway, perilacunar remodelling, type of fixation and putative biomarkers to monitor fracture healing are highly desirable to bridge the current gaps of knowledge.The translational potential of this article: This systematic review provides an up-to-date chronological overview and highlights the osteocyte-regulated events at gene, protein, cellular and tissue levels throughout the fracture healing cascade, with the hope of informing and developing potential new therapeutic strategies that could improve the timing and quality of fracture healing in the future.
Collapse
Affiliation(s)
- Man Huen Victoria Choy
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, PR China
| | - Ronald Man Yeung Wong
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, PR China
| | - Simon Kwoon Ho Chow
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, PR China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
| | - Meng Chen Li
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, PR China
| | - Yu Ning Chim
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, PR China
| | - Tsz Kiu Li
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, PR China
| | - Wing Tung Ho
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, PR China
| | - Jack Chun Yiu Cheng
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, PR China
| | - Wing Ho Cheung
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, PR China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China
- Corresponding author. Department of Orthopaedics and Traumatology, 5/F, Lui Che Woo Clinical Sciences Building, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, PR China.
| |
Collapse
|
11
|
Nakamura S, Sato Y, Kobayashi T, Oike T, Kaneko Y, Miyamoto K, Funayama A, Oya A, Nishiwaki T, Matsumoto M, Nakamura M, Kanaji A, Miyamoto T. Insulin-like growth factor-I is required to maintain muscle volume in adult mice. J Bone Miner Metab 2019; 37:627-635. [PMID: 30324536 DOI: 10.1007/s00774-018-0964-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/25/2018] [Indexed: 12/20/2022]
Abstract
Insulin-like growth factor-I (IGF-I) is a peptide with diverse functions, among them regulation of embryonic development and bone homeostasis. Serum IGF-I levels decline in the elderly; however, IGF-I function in adults has not been clearly defined. Here, we show that IGF-I is required to maintain muscle mass in adults. We crossed Igf-I flox'd and Mx1 Cre mice to yield Mx1 Cre/Igf-Iflox/flox (IGF-I cKO) mice, and deleted Igf-I in adult mice by polyIpolyC injection. We demonstrate that, although serum IGF-I levels significantly decreased after polyIpolyC injection relative to (Igf-Iflox/flox) controls, serum glucose levels were unchanged. However, muscle mass decreased significantly after IGF-I down-regulation, while bone mass remained the same. In IGF-I cKO muscle, expression of anabolic factors such as Eif4e and p70S6K significantly decreased, while expression of catabolic factors MuRF1 and Atrogin-1 was normal and down-regulated, respectively, suggesting that observed muscle mass reduction was due to perturbed muscle metabolism. Our data demonstrate a specific role for IGF-I in maintaining muscle homeostasis in adults.
Collapse
Affiliation(s)
- Satoshi Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuiko Sato
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Advanced Therapy for Musculoskeletal Disorders, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Tami Kobayashi
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Musculoskeletal Reconstruction and Regeneration Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takatsugu Oike
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yosuke Kaneko
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kana Miyamoto
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Atsushi Funayama
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Akihito Oya
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Toru Nishiwaki
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Morio Matsumoto
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Arihiko Kanaji
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Takeshi Miyamoto
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan.
- Department of Musculoskeletal Reconstruction and Regeneration Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
12
|
Liu H, Su H, Wang X, Hao W. MiR-148a regulates bone marrow mesenchymal stem cells-mediated fracture healing by targeting insulin-like growth factor 1. J Cell Biochem 2019; 120:1350-1361. [PMID: 30335895 DOI: 10.1002/jcb.27121] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/04/2018] [Indexed: 01/24/2023]
Abstract
The purpose of this study was to investigate the underlying molecular mechanisms of fracture healing mediated by bone marrow mesenchymal stem cells. Differentially expressed microRNAs in acutely injured subjects and healthy volunteers were screened by microarray analysis. The dual luciferase reporter system was used to verify whether insulin-like growth factor 1 (IGF1) was the direct target gene regulated by miR-148a. The expression level of miR-148a and IGF1 after osteogenic differentiation was detected by quantitative real-time polymerase chain reaction. Western blot was used to determine the protein expression of bone markers, including IGF1, runt-related transcription factor 2 (Runx2), osteocalcin, and osteopontin in rat bone marrow-derived mesenchymal stem cells. Alkaline phosphatase and alizarin red staining was used to detect alkaline phosphatase activity and calcium deposition. An animal fracture model was used for in vivo experiments. MiR-148a was highly expressed in acutely injured subjects compared with healthy volunteers, and IGF1 was a target of miR-148a. Moreover, compared with the negative control group, IGF1 messenger RNA expression was significantly increased in the miR-148a antagomir group. During osteogenic differentiation, the expression of IGF1, Runx2, osteocalcin, and osteopontin was higher in the miR-148a antagomir group than other groups. In vivo experiments further confirmed that upregulation of IGF1 enhanced fracture healing efficiently by decreasing callus width and area and improving bone mineral density, maximum load, stiffness, and energy absorption. It was proved that IGF1 was the direct target gene of miR-148a, and the use of rat bone marrow-derived mesenchymal stem cells with low expression of miR-148a could improve fracture healing by upregulating IGF1.
Collapse
Affiliation(s)
- Hongzhi Liu
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Hao Su
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Xin Wang
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Wei Hao
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| |
Collapse
|
13
|
Gao X, Lu A, Tang Y, Schneppendahl J, Liebowitz AB, Scibetta AC, Morris ER, Cheng H, Huard C, Amra S, Wang B, Hall MA, Lowe WR, Huard J. Influences of donor and host age on human muscle-derived stem cell-mediated bone regeneration. Stem Cell Res Ther 2018; 9:316. [PMID: 30463597 PMCID: PMC6249775 DOI: 10.1186/s13287-018-1066-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/14/2018] [Accepted: 11/05/2018] [Indexed: 01/08/2023] Open
Abstract
Background Human muscle-derived stem cells (hMDSCs) have been shown to regenerate bone efficiently when they were transduced with Lenti-viral bone morphogenetic protein 2 (LBMP2). However, whether the age of hMDSCs and the animal host affect the bone regeneration capacity of hMDSCs and mechanism are unknown which prompted the current study. Methods We isolated three gender-matched young and old populations of skeletal muscle stem cells, and tested the influence of cells’ age on in vitro osteogenic differentiation using pellet culture before and after Lenti-BMP2/green fluorescent protein (GFP) transduction. We further investigated effects of the age of hMDSCs and animal host on hMDSC-mediated bone regeneration in a critical-size calvarial bone defect model in vivo. Micro-computer tomography (CT), histology, and immunohistochemistry were used to evaluate osteogenic differentiation and mineralization in vitro and bone regeneration in vivo. Western blot, quantitative polymerase chain reaction (PCR), and oxidative stress assay were performed to detect the effects of age of hMDSCs on cell survival and osteogenic-related genes. Serum insulin-like growth factor 1 (IGF1) and receptor activator of nuclear factor-kappa B ligand (RANKL) were measured with an enzyme-linked immunosorbent assay (ELISA). Results We found LBMP2/GFP transduction significantly enhanced osteogenic differentiation of hMDSCs in vitro, regardless of donor age. We also found old were as efficient as young LBMP2/GFP-transduced hMDSCs for regenerating functional bone in young and old mice. These findings correlated with lower phosphorylated p38MAPK expression and similar expression levels of cell survival genes and osteogenic-related genes in old hMDSCs relative to young hMDSCs. Old cells exhibited equivalent resistance to oxidative stress. However, both young and old donor cells regenerated less bone in old than young hosts. Impaired bone regeneration in older hosts was associated with high bone remodeling due to higher serum levels of RANKL and lower level of IGF-1. Conclusion hMDSC-mediated bone regeneration was not impaired by donor age when hMDSCs were transduced with LBMP2/GFP, but the age of the host adversely affected hMDSC-mediated bone regeneration. Regardless of donor and host age, hMDSCs formed functional bone, suggesting a promising cell resource for bone regeneration. Electronic supplementary material The online version of this article (10.1186/s13287-018-1066-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xueqin Gao
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA.,Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA.,Steadman Philippon Research Institute, Vail, CO, 81657, USA
| | - Aiping Lu
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA.,Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA.,Steadman Philippon Research Institute, Vail, CO, 81657, USA
| | - Ying Tang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | | | | | | | | | - Haizi Cheng
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Charles Huard
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Sarah Amra
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Bing Wang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Mary A Hall
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Walter R Lowe
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA. .,Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77054, USA. .,Steadman Philippon Research Institute, Vail, CO, 81657, USA.
| |
Collapse
|
14
|
Ding ZC, Lin YK, Gan YK, Tang TT. Molecular pathogenesis of fracture nonunion. J Orthop Translat 2018; 14:45-56. [PMID: 30035032 PMCID: PMC6019407 DOI: 10.1016/j.jot.2018.05.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 02/07/2023] Open
Abstract
Fracture nonunion, a serious bone fracture complication, remains a challenge in clinical practice. Although the molecular pathogenesis of nonunion remains unclear, a better understanding may provide better approaches for its prevention, diagnosis and treatment at the molecular level. This review tries to summarise the progress made in studies of the pathogenesis of fracture nonunion. We discuss the evidence supporting the concept that the development of nonunion is related to genetic factors. The importance of several cytokines that regulate fracture healing in the pathogenesis of nonunion, such as tumour necrosis factor-α, interleukin-6, bone morphogenetic proteins, insulin-like growth factors, matrix metalloproteinases and vascular endothelial growth factor, has been proven in vitro, in animals and in humans. Nitric oxide and the Wnt signalling pathway also play important roles in the development of nonunion. We present potential strategies for the prevention, diagnosis and treatment of nonunion, and the interaction between genetic alteration and abnormal cytokine expression warrants further investigation. The translational potential of this article A better understanding of nonunion molecular pathogenesis may provide better approaches for its prevention, diagnosis and treatment in clinical practice.
Collapse
Affiliation(s)
- Zi-Chuan Ding
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, 639 Zhizaoju Road, Shanghai, China
| | - Yi-Kai Lin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, 639 Zhizaoju Road, Shanghai, China
| | - Yao-Kai Gan
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, 639 Zhizaoju Road, Shanghai, China
| | - Ting-Ting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, 639 Zhizaoju Road, Shanghai, China
| |
Collapse
|
15
|
Fan T, Huang G, Wu W, Guo R, Zeng Q. Combined treatment with extracorporeal shock‑wave therapy and bone marrow mesenchymal stem cell transplantation improves bone repair in a rabbit model of bone nonunion. Mol Med Rep 2017; 17:1326-1332. [PMID: 29115642 DOI: 10.3892/mmr.2017.7984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 06/08/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to analyze whether extracorporeal shock‑wave therapy (ESWT) combined with bone marrow mesenchymal stem cell (BMMSC) transplantation improves bone repair in a rabbit bone nonunion model. ESWT combined with BMMSC effectively enhanced mechanical strength, fracture stiffness and histological scores, and increased alkaline phosphatase activity, and osteopontin, runt related transcription factor 2 and collagen type I α1 chain protein expression levels in a rabbit bone nonunion model. In addition, ESWT combined with BMMSC effectively enhanced insulin‑like growth factor 1 and vascular endothelial growth factor contents, promoted transforming growth factor‑β (TGF‑β) contents, and induced the growth factors, bone morphogenetic protein (BMP)‑2, BMP‑4 and purinergic receptor P2X7 (P2X7) protein expression in the rabbit bone nonunion model. Thus, the present study demonstrated that ESWT combined with BMMSC transplantation improves bone repair in a rabbit bone nonunion model via the BMPs and P2X7 signaling pathways.
Collapse
Affiliation(s)
- Tao Fan
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Guozhi Huang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Wen Wu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Rong Guo
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Qing Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
16
|
Abstract
When normal physiologic functions go awry, disorders and disease occur. This is universal; even for the osteocyte, a cell embedded within the mineralized matrix of bone. It was once thought that this cell was simply a placeholder in bone. Within the last decade, the number of studies of osteocytes has increased dramatically, leading to the discovery of novel functions of these cells. With the discovery of novel physiologic functions came the discoveries of how these cells can also be responsible for not only bone diseases and disorders, but also those of the kidney, heart, and potentially muscle.
Collapse
Affiliation(s)
- Lynda F Bonewald
- Indiana Center for Musculoskeletal Health, VanNuys Medical Science Building, MS 5055, 635 Barnhill Drive, Indianapolis, IN 46202, USA; Department of Anatomy and Cell Biology, VanNuys Medical Science Building, MS 5035, Indianapolis, IN 46202, USA; Department of Orthopaedic Surgery, 1120 West Michigan Street, Suite 600, Indianapolis, IN 46202, USA.
| |
Collapse
|