1
|
Wu J, Li R, Liu C, Li W. The role of AKR1B10 in osteogenic differentiation of mesenchymal stem cells and atrophic nonunion. Bone 2024; 190:117284. [PMID: 39401534 DOI: 10.1016/j.bone.2024.117284] [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: 08/26/2024] [Revised: 09/29/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Atrophic nonunion is a chronic disease without effective medications. Here, high-throughput mRNA sequencing was used to explore the novel targets in atrophic nonunion. AKR1B10, a member of aldo-keto reductase family 1, is upregulated in atrophic nonunion tissues. There are currently no studies to reveal the role of AKR1B10 in atrophic nonunion. We used rat bone marrow-derived mesenchymal stem cells (BMSCs) to explore the effect of AKR1B10 on the osteogenic differentiation and autophagy. In vivo, we implanted collagen sponges loaded with LV-shAKR1B10-transduced BMSCs into rat fractured femurs to explore the role of AKR1B10 in fracture healing. The results showed that AKR1B10 reduced the activity of ALP, suppressed the expression of COL1A1, RUNX2 and OCN, and inhibited calcification deposition in osteogenically differentiated BMSCs. AKR1B10 reduced the expression of LC3II, decreased the number of autophagosomes, and promoted the expression of p62. In addition, the promoting effect of AKR1B10 knockdown on osteogenic differentiation of BMSCs was attenuated by 3-MA treatment. Implantation of collagen sponges found that knockdown of AKR1B10 promoted bone fracture healing. In conclusion, AKR1B10 inhibited the osteogenic differentiation and autophagy, and delayed the bone fracture healing. These results provide a new perspective on revealing the role of AKR1B10 in nonunion and may also provide a new therapeutic target for the treatment of nonunion.
Collapse
Affiliation(s)
- Jie Wu
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Runze Li
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Chen Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Weiming Li
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
2
|
Mahajan A, Bhattacharyya S. Immunomodulation by mesenchymal stem cells during osteogenic differentiation: Clinical implications during bone regeneration. Mol Immunol 2023; 164:143-152. [PMID: 38011783 DOI: 10.1016/j.molimm.2023.11.006] [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: 06/12/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 11/29/2023]
Abstract
Critical bone defects resulting in delayed and non-union are a major concern in the field of orthopedics. Over the past decade, mesenchymal stem cells (MSCs) have become a promising frontier for bone repair and regeneration owing to their high expansion rate and osteogenic differentiation potential ex vivo. MSCs have also long been associated with their ability to modulate immune response in the recipients. These can even skew the immune response towards pro-inflammatory or anti-inflammatory type by sensing their local microenvironment. MSCs adopt anti-inflammatory phenotype at bone injury site and secrete various immunomodulatory factors such as IDO, NO, TGFβ1 and PGE-2 which have redundant role in osteoblast differentiation and bone formation. As such, several studies have also sought to decipher the immunomodulatory effects of osteogenically differentiated MSCs. The present review discusses the immunomodulatory status of MSCs during their osteogenic differentiation and summarizes few mechanisms that cause immunosuppression by osteogenically differentiated MSCs and its implication during bone healing.
Collapse
Affiliation(s)
- Aditi Mahajan
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shalmoli Bhattacharyya
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
3
|
Smolinska V, Csobonyeiova M, Zamborsky R, Danisovic L. Stem Cells and Their Derivatives: An Implication for the Regeneration of Nonunion Fractures. Cell Transplant 2023; 32:9636897231183530. [PMID: 37462248 PMCID: PMC10363876 DOI: 10.1177/09636897231183530] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 07/20/2023] Open
Abstract
Despite advances in biomedical research, fracture nonunion rates have remained stable throughout the years. Long-bone fractures have a high likelihood of nonunion, but the specific biological pathways involved in this severe consequence are unknown. Fractures often heal in an organized sequence, including the production of a hematoma and an early stage of inflammation, the development of a soft callus and hard callus, and eventually the stage of bone remodeling. Deficient healing can result in a persistent bone defect with instability, discomfort, and loss of function. In the treatment of nonunions, mesenchymal stem cells (MSCs) prove to be a promising and safe alternative to the standard therapeutic strategies. Moreover, novel scaffolds are being created in order to use a synergistic biomimetic technique to rapidly generate bone tissue. MSCs respond to acellular biomimetic matrices by regenerating bone. Extracellular vesicles (EVs) derived from MSCs have recently gained interest in the field of musculoskeletal regeneration. Although many of these techniques and technologies are still in the preclinical stage and have not yet been approved for use in humans, novel approaches to accelerate bone healing via MSCs and/or MSC derivatives have the potential to reduce the physical, economic, and social burdens associated with nonhealing fractures and bone defects. In this review, we focus on providing an up-to-date summary of recent scientific studies dealing with the treatment of nonunion fractures in clinical and preclinical settings employing MSC-based therapeutic techniques.
Collapse
Affiliation(s)
- Veronika Smolinska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
- National Institute of Rheumatic Diseases, Piestany, Slovakia
| | - Maria Csobonyeiova
- National Institute of Rheumatic Diseases, Piestany, Slovakia
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Radoslav Zamborsky
- National Institute of Rheumatic Diseases, Piestany, Slovakia
- Department of Orthopaedics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
- National Institute of Children’s Diseases, Bratislava, Slovakia
- Centre for Tissue Engineering and Regenerative Medicine–Translational Research Unit, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
- National Institute of Rheumatic Diseases, Piestany, Slovakia
- Centre for Tissue Engineering and Regenerative Medicine–Translational Research Unit, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
4
|
Menger MM, Laschke MW, Nussler AK, Menger MD, Histing T. The vascularization paradox of non-union formation. Angiogenesis 2022; 25:279-290. [PMID: 35165821 PMCID: PMC9249698 DOI: 10.1007/s10456-022-09832-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/21/2021] [Indexed: 01/01/2023]
Abstract
Despite major research efforts to elucidate mechanisms of non-union formation, failed fracture healing remains a common complication in orthopedic surgery. Adequate vascularization has been recognized as a crucial factor for successful bone regeneration, as newly formed microvessels guarantee the supply of the callus tissue with vital oxygen, nutrients, and growth factors. Accordingly, a vast number of preclinical studies have focused on the development of vascularization strategies to stimulate fracture repair. However, recent evidence suggests that stimulation of blood vessel formation is an oversimplified approach to support bone regeneration. This review discusses the role of vascularization during bone regeneration and delineates a phenomenon, for which we coin the term "the vascularization paradox of non-union-formation". This view is based on the results of a variety of experimental studies that suggest that the callus tissue of non-unions is indeed densely vascularized and that pro-angiogenic mediators, such as vascular endothelial growth factor, are sufficiently expressed at the facture site. By gaining further insights into the molecular and cellular basis of non-union vascularization, it may be possible to develop more optimized treatment approaches or even prevent the non-union formation in the future.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany.
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany.
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Andreas K Nussler
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Tina Histing
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| |
Collapse
|
5
|
Sabbaghzadeh A, Bonakdar S, Gorji M, Gholipour M. Evaluation of the effect of preoperative hemoglobin level and proinflammatory factors on intertrochanteric fracture union. Wien Klin Wochenschr 2022; 134:458-462. [PMID: 35639200 DOI: 10.1007/s00508-022-02042-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Intertrochanteric fractures are associated with high mortality and morbidity, so these patients should undergo fracture fixation surgery immediately. Despite surgery, the possibility of fracture fusion may not occur due to the association with various causes. Therefore, our aim is to investigate these factors (TNF‑a, IL‑1, Hb) and their effect on fracture union after fixation. METHODS From 2018 to 2020, at our orthopedic trauma center, 163 patients older than 50 years with intertrochanteric fractures underwent DHS fixation surgery. Patients were divided into anemic and non-anemic groups in terms of preoperative hemoglobin level (standard hemoglobin 11 mg/dl). For 3 months, patients were assessed for union and failure fixation criteria, levels of proinflammation (TNF‑α, IL-1) and level of hemoglobin. RESULTS The results show that out of 163 patients with fractures, at the time of initial admission, 74 patients had less than 11 hemoglobin g/dl. Patients with union fractures had higher hemoglobin levels than patients with non-union (11.71 ± 1.51 versus 11.24 ± 1.96), which was statistically significant between hemoglobin and union level (p = 0.030). At the end of the third visit (third month), 44 (59.5%) anemic patients received union completly, while among the patients with normal hemoglobin level, 32 (36%) received union bread, which was statistically significant (p = 0.003). There were no statistically significant differences between proinflammatory factors before surgery and 3 months after surgery (p > 0.05). CONCLUSION Due to the effect of anemia and proinflammatory factors in the process of healing fractures and bone formation and creating musculoskeletal balance, low hemoglobin level before surgery has a significant effect on fracture union and failure of fixation. So it is recommended to correct this anemia in these patients before surgery and during follow-up.
Collapse
Affiliation(s)
- Amir Sabbaghzadeh
- Physiotherapy Research Center, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran.,Clinical Research Development Unit, Akhtar Hospital Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Sona Bonakdar
- Department of Foreign Languages, Urmia University, Urmia, Iran
| | - Mona Gorji
- Physiotherapy Research Center, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran.,Skin research center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Morteza Gholipour
- Physiotherapy Research Center, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran. .,Clinical Research Development Unit, Akhtar Hospital Shahid Beheshti University of Medical Science, Tehran, Iran.
| |
Collapse
|
6
|
Thurairajah K, Briggs GD, Balogh ZJ. Stem cell therapy for fracture non-union: The current evidence from human studies. J Orthop Surg (Hong Kong) 2021; 29:23094990211036545. [PMID: 34396805 DOI: 10.1177/23094990211036545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Non-union is a taxing complication of fracture management for both the patient and their surgeon. Modern fracture fixation techniques have been developed to optimise the biomechanical environment for fracture healing but do not guarantee union. Patient biology has a critical role in achieving union and stem cell therapy has potential for improving fracture healing at a cellular level to treat or avoid non-union. This article reviews the current understanding of non-union, concepts in bone healing and the current literature on the application of stem cells in non-union.
Collapse
Affiliation(s)
- Kabilan Thurairajah
- Department of Traumatology, 37024John Hunter Hospital and University of Newcastle, Newcastle, Australia
| | - Gabrielle D Briggs
- School of Medicine and Public Health, 5982University of Newcastle, Newcastle, Australia
| | - Zsolt J Balogh
- Department of Traumatology, 37024John Hunter Hospital and University of Newcastle, Newcastle, Australia
| |
Collapse
|
7
|
Sadeghifar A, Sheibani M, Joukar S, Dabiri S, Alavi S, Azari O, Vosoghi D, Zeynali Y, Zeynali Y, Shahraki M, Torghabe A, Rostamzadeh F, Nasri A. The Effect of Waterpipe Tobacco Smoking on Bone Healing Following Femoral Fractures in Male Rats. Front Surg 2021; 8:722446. [PMID: 34671637 PMCID: PMC8520932 DOI: 10.3389/fsurg.2021.722446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/30/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Given the increasing use of waterpipe tobacco smoking in the world and its unknown effects on bone healing, this study investigated the repairing of femoral bone fractures in rats exposed to waterpipe tobacco smoking (WTS). Main Methods: This study involved 40 male Wistar rats that were divided into two groups, including the femoral fracture (Fx) and the Fx + WTS groups. Each group was divided into two subgroups that were evaluated for bone healing 28 and 42 days after femoral fracture. After fixing the fractured femur, the healing process was evaluated by radiography, pathological indicators, and a measurement of the blood levels of vascular endothelial growth factor (VEGF), parathyroid hormone (PTH), Ca ++, transforming growth factor-beta (TGF-β), and insulin-like growth factor 1 (IGF-1). Additionally, the density of VEGF and CD34 in fracture tissue was investigated by immunohistochemistry. Key Findings: Radiographic findings showed that factors related to the earlier stages of bone healing had higher scores in the Fx + WTS28 and 42 subgroups in comparison to the Fx groups. The density of VEGF and CD34 showed that the angiogenesis processes were different in the bone fracture area and callus tissue in the Fx +WTS subgroups. The serum levels of VEGF, TGF-β, and IGF-1 were significantly lower in the Fx +WTS42 group, and PTH in the Fx +WTS28 group was higher than that in the other groups. Significance: The findings showed the disturbance and delay in the femoral fracture union in rats exposed to hookah smoke. This is partly due to the reduction of molecular stimuli of bone synthesis and the attenuation of quantitative angiogenesis.
Collapse
Affiliation(s)
- Amirreza Sadeghifar
- Orthopedic Department, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohamad Sheibani
- Orthopedic Department, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Siyavash Joukar
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, and Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Pathology Department and Stem Cell Research Center, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Samanehsadat Alavi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Omid Azari
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Darioush Vosoghi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Yas Zeynali
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, and Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Yasman Zeynali
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, and Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohamad Shahraki
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Amirhesam Torghabe
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Farzaneh Rostamzadeh
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Nasri
- Pathology Department and Stem Cell Research Center, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
8
|
A Chemotactic Functional Scaffold with VEGF-Releasing Peptide Amphiphiles Facilitates Bone Regeneration by BMP-2 in a Large-Scale Rodent Cranial Defect Model. Plast Reconstr Surg 2021; 147:386-397. [PMID: 33235044 DOI: 10.1097/prs.0000000000007551] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Current common techniques for repairing calvarial defects by autologous bone grafting and alloplastic implants have significant limitations. In this study, the authors investigated a novel alternative approach to bone repair based on peptide amphiphile nanofiber gels that are engineered to control the release of vascular endothelial growth factor (VEGF) to recruit circulating stem cells to a site of bone regeneration and facilitate bone healing by bone morphogenetic protein-2 (BMP-2). METHODS VEGF release kinetics from peptide amphiphile gels were evaluated. Chemotactic functional scaffolds were fabricated by combining collagen sponges with peptide amphiphile gels containing VEGF. The in vitro and in vivo chemotactic activities of the scaffolds were evaluated by measuring mesenchymal stem cell migration, and angiogenic capability of the scaffolds was also evaluated. Large-scale rodent cranial bone defects were created to evaluate bone regeneration after implanting the scaffolds and other control materials. RESULTS VEGF was released from peptide amphiphile in a controlled-release manner. In vitro migration of mesenchymal stem cells was significantly greater when exposed to chemotactic functional scaffolds compared to control scaffolds. In vivo chemotaxis was evidenced by migration of tracer-labeled mesenchymal stem cells to the chemotactic functional scaffolds. Chemotactic functional scaffolds showed significantly increased angiogenesis in vivo. Successful bone regeneration was noted in the defects treated with chemotactic functional scaffolds and BMP-2. CONCLUSIONS The authors' observations suggest that this bioengineered construct successfully acts as a chemoattractant for circulating mesenchymal stem cells because of controlled release of VEGF from the peptide amphiphile gels. The chemotactic functional scaffolds may play a role in the future design of clinically relevant bone graft substitutes for large-scale bone defects.
Collapse
|
9
|
Wu P, Zhang X, Hu Y, Liu D, Song J, Xu W, Tan H, Lu R, Zheng L. Co-culture with Endothelial Progenitor Cells promotes the Osteogenesis of Bone Mesenchymal Stem Cells via the VEGF-YAP axis in high-glucose environments. Int J Med Sci 2021; 18:1628-1638. [PMID: 33746579 PMCID: PMC7976568 DOI: 10.7150/ijms.52316] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 01/11/2021] [Indexed: 11/05/2022] Open
Abstract
Patients with type 2 diabetes mellitus (T2DM) have a high risk of fracture and experience poor bone healing. In recent years, bone mesenchymal stem cells (BMSCs) and endothelial progenitor cells (EPCs) have become the most commonly used cells in cell therapy and tissue engineering. In this study, we found that high glucose levels had a negative effect on the differentiation of BMSCs and EPCs. Considering that EPCs-BMSCs sheets can provide endothelial cells and osteoblastic cells, we transplanted cell sheets into T2DM rats with bilateral skull defects. The outcomes of the in vivo study revealed that EPCs-BMSCs sheets promoted ossification, which was verified by micro-CT and immunohistochemistry (IHC) analyses. Furthermore, we detected the VEGF content in the culture supernatant using an enzyme-linked immunosorbent assay (ELISA). The results showed that the BMSCs co-cultured with EPCs presented a higher level of VEGF than other cells. To assess the differentiation and migration of BMSCs exposed to VEGF, ALP staining, scratch assay and qRT-PCR analysis were performed. In addition, we used immunofluorescence and western blotting analysis to further explore the related mechanisms. The results showed that cells cultured with VEGF had a stronger actin cytoskeleton and a greater amount of nuclear and total YAP than cells cultured without VEGF. Taken together, our results indicate that co-culture with EPCs could promote the osteogenesis of BMSCs partially via VEGF. Furthermore, YAP and F-actin play important roles in this process.
Collapse
Affiliation(s)
- Peilian Wu
- The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Xia Zhang
- The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, 401147, China
- West china dental hospital of Chongqing, Chongqing, 401147, China
| | - Yun Hu
- The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Dongrong Liu
- The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Jinlin Song
- The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Wenjie Xu
- The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Hao Tan
- The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Rui Lu
- The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Leilei Zheng
- The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| |
Collapse
|
10
|
Choi D, Heo J, Aviles Milan J, Oreffo ROC, Dawson JI, Hong J, Kim YH. Structured nanofilms comprising Laponite® and bone extracellular matrix for osteogenic differentiation of skeletal progenitor cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 118:111440. [PMID: 33255033 DOI: 10.1016/j.msec.2020.111440] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 08/16/2020] [Accepted: 08/22/2020] [Indexed: 12/11/2022]
Abstract
Functionalized scaffolds hold promise for stem cell therapy by controlling stem cell fate and differentiation potential. Here, we have examined the potential of a 2-dimensional (2D) scaffold to stimulate bone regeneration. Solubilized extracellular matrix (ECM) from human bone tissue contains native extracellular cues for human skeletal cells that facilitate osteogenic differentiation. However, human bone ECM displays limited mechanical strength and degradation stability under physiological conditions, necessitating modification of the physical properties of ECM before it can be considered for tissue engineering applications. To increase the mechanical stability of ECM, we explored the potential of synthetic Laponite® (LAP) clay as a counter material to prepare a 2D scaffold using Layer-by-Layer (LbL) self-assembly. The LAP and ECM multilayer nanofilms (ECM/LAP film) were successfully generated through electrostatic and protein-clay interactions. Furthermore, to enhance the mechanical properties of the ECM/LAP film, application of a NaCl solution wash step, instead of deionized water following LAP deposition resulted in the generation of stable, multi-stacked LAP layers which displayed enhanced mechanical properties able to sustain human skeletal progenitor cell growth. The ECM/LAP films were not cytotoxic and, critically, showed enhanced osteogenic differentiation potential as a consequence of the synergistic effects of ECM and LAP. In summary, we demonstrate the fabrication of a novel ECM/LAP nanofilm layer material with potential application in hard tissue engineering.
Collapse
Affiliation(s)
- Daheui Choi
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Jiwoong Heo
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Juan Aviles Milan
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, SO16 6YD, United Kingdom
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, SO16 6YD, United Kingdom
| | - Jonathan I Dawson
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, SO16 6YD, United Kingdom.
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea.
| | - Yang-Hee Kim
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, SO16 6YD, United Kingdom.
| |
Collapse
|
11
|
Xie H, Liu M, Jin Y, Lin H, Zhang Y, Zheng S. miR-1323 suppresses bone mesenchymal stromal cell osteogenesis and fracture healing via inhibiting BMP4/SMAD4 signaling. J Orthop Surg Res 2020; 15:237. [PMID: 32600409 PMCID: PMC7322887 DOI: 10.1186/s13018-020-01685-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/29/2020] [Indexed: 12/14/2022] Open
Abstract
Background Atrophic non-union fractures show no radiological evidence of callus formation within 3 months of fracture. microRNA dysregulation may underlie the dysfunctional osteogenesis in atrophic non-union fractures. Here, we aimed to analyze miR-1323 expression in human atrophic non-union fractures and examine miR-1323’s underlying mechanism of action in human mesenchymal stromal cells. Methods Human atrophic non-union and standard healing fracture specimens were examined using H&E and Alcian Blue staining, immunohistochemistry, qRT-PCR, immunoblotting, and ALP activity assays. The effects of miR-1323 mimics or inhibition on BMP4, SMAD4, osteogenesis-related proteins, ALP activity, and bone mineralization were analyzed in human mesenchymal stromal cells. Luciferase reporter assays were utilized to assay miR-1323’s binding to the 3'UTRs of BMP4 and SMAD4. The effects of miR-1323, BMP4, and SMAD4 were analyzed by siRNA and overexpression vectors. A rat femur fracture model was established to analyze the in vivo effects of antagomiR-1323 treatment. Results miR-1323 was upregulated in human atrophic non-union fractures. Atrophic non-union was associated with downregulation of BMP4 and SMAD4 as well as the osteogenic markers ALP, collagen I, and RUNX2. In vitro, miR-1323 suppressed BMP4 and SMAD4 expression by binding to the 3'UTRs of BMP4 and SMAD4. Moreover, miR-1323’s inhibition of BMP4 and SMAD4 inhibited mesenchymal stromal cell osteogenic differentiation via modulating the nuclear translocation of the transcriptional co-activator TAZ. In vivo, antagomiR-1323 therapy facilitated the healing of fractures in a rat model of femoral fracture. Conclusions This evidence supports the miR-1323/BMP4 and miR-1323/SMAD4 axes as novel therapeutic targets for atrophic non-union fractures.
Collapse
Affiliation(s)
- Hui Xie
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Ming Liu
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Yaofeng Jin
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Haiqing Lin
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Yushan Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Song Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China.
| |
Collapse
|
12
|
LncRNA ENST00000563492 promoting the osteogenesis-angiogenesis coupling process in bone mesenchymal stem cells (BMSCs) by functions as a ceRNA for miR-205-5p. Cell Death Dis 2020; 11:486. [PMID: 32587236 PMCID: PMC7316863 DOI: 10.1038/s41419-020-2689-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022]
Abstract
Pain, physical dysfunction, and mental disorders caused by bone nonunion bring great burden to patients. Bone mesenchymal stem cells (BMSCs) isolated from bone nonunion patients with poor proliferation and osteogenic ability are compared with that from normal bone-healing patients. Long noncoding RNAs (lncRNAs) are a class of RNAs that are more than 200 nucleotides in length, lack an open-reading frame encoding a protein, and have little or no protein-coding function, and could regulate gene expression, which is involved in the regulation of important life activities, such as growth, development, aging, and death at epigenetic, transcriptional, and post-transcriptional levels. In this study, we intended to investigate the difference of lncRNA expression between patients with nonunion and normal fracture healing. Our result found that lncRNA ENST00000563492 was downregulated in bone nonunion tissues. LncRNA ENST00000563492 promotes osteogenic differentiation of BMSCs through upregulating the expression of CDH11. On the other hand, LncRNA ENST0000563492 could improve the osteogenesis–angiogenesis coupling process through enhancing the expression of VEGF during osteogenic differentiation of BMSCs. LncRNA ENST00000563492 functions as a ceRNA for miR-205-5p that was targeting CDH11 and VEGF. LncRNA ENST00000563492 could promote the osteogenesis of BMSCs in vivo. Our result indicated that lncRNA ENST00000563492 may be a new target for bone nonunion.
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW The failure of bony union following a fracture, termed a fracture nonunion, has severe patient morbidity and economic consequences. This review describes current consensuses and future directions of investigation for determining why, detecting when, and effective treatment if this complication occurs. RECENT FINDINGS Current nonunion investigation is emphasizing an expanded understanding of the biology of healing. This has led to assessments of the immune environment, multiple cytokines and morphogenetic factors, and the role of skeletogenic stem cells in the development of nonunion. Detecting biological markers and other objective diagnostic criteria is also a current objective of nonunion research. Treatment approaches in the near future will likely be dominated by the development of specific adjunct therapies to the nonunion surgical management, which will be informed by an expanded mechanistic understanding of nonunion biology. Current consensus among orthopedists is that improved diagnosis and treatment of nonunion hinges first on discoveries at the bench side with later translation to the clinic.
Collapse
Affiliation(s)
- G Bradley Reahl
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, 02118, USA.
| | - Louis Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Michael Kain
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
14
|
Lienemann PS, Vallmajo‐Martin Q, Papageorgiou P, Blache U, Metzger S, Kiveliö A, Milleret V, Sala A, Hoehnel S, Roch A, Reuten R, Koch M, Naveiras O, Weber FE, Weber W, Lutolf MP, Ehrbar M. Smart Hydrogels for the Augmentation of Bone Regeneration by Endogenous Mesenchymal Progenitor Cell Recruitment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903395. [PMID: 32274319 PMCID: PMC7141038 DOI: 10.1002/advs.201903395] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/27/2019] [Indexed: 04/14/2023]
Abstract
The treatment of bone defects with recombinant bone morphogenetic protein-2 (BMP-2) requires high doses precluding broad clinical application. Here, a bioengineering approach is presented that strongly improves low-dose BMP-2-based bone regeneration by mobilizing healing-associated mesenchymal progenitor cells (MPCs). Smart synthetic hydrogels are used to trap and study endogenous MPCs trafficking to bone defects. Hydrogel-trapped and prospectively isolated MPCs differentiate into multiple lineages in vitro and form bone in vivo. In vitro screenings reveal that platelet-derived growth factor BB (PDGF-BB) strongly recruits prospective MPCs making it a promising candidate for the engineering of hydrogels that enrich endogenous MPCs in vivo. However, PDGF-BB inhibits BMP-2-mediated osteogenesis both in vitro and in vivo. In contrast, smart two-way dynamic release hydrogels with fast-release of PDGF-BB and sustained delivery of BMP-2 beneficially promote the healing of bone defects. Collectively, it is shown that modulating the dynamics of endogenous progenitor cells in vivo by smart synthetic hydrogels significantly improves bone healing and holds great potential for other advanced applications in regenerative medicine.
Collapse
Affiliation(s)
- Philipp S. Lienemann
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Queralt Vallmajo‐Martin
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Panagiota Papageorgiou
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
| | - Ulrich Blache
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
| | - Stéphanie Metzger
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Anna‐Sofia Kiveliö
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Vincent Milleret
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
| | - Ana Sala
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
| | - Sylke Hoehnel
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Aline Roch
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Raphael Reuten
- Institute for Dental Research and Oral Musculoskeletal BiologyCenter for BiochemistryUniversity of CologneCologne50931Germany
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal BiologyCenter for BiochemistryUniversity of CologneCologne50931Germany
| | - Olaia Naveiras
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Franz E. Weber
- Department of Cranio‐Maxillofacial SurgeryOral Biotechnology and BioengineeringUniversity Hospital ZurichFrauenklinikstrasse 24Zurich8091Switzerland
| | - Wilfried Weber
- Faculty of Biology and BIOSS Centre for Biological Signalling StudiesUniversity of FreiburgSchänzlestr. 18Freiburg79104Germany
| | - Matthias P. Lutolf
- Institute of BioengineeringSchool of Life Sciences and School of EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL)Station 15Lausanne1015Switzerland
| | - Martin Ehrbar
- Department of ObstetricsUniversity Hospital ZurichUniversity of ZurichSchmelzbergstr. 12Zurich8091Switzerland
| |
Collapse
|
15
|
Zhang L, Jiao G, Ren S, Zhang X, Li C, Wu W, Wang H, Liu H, Zhou H, Chen Y. Exosomes from bone marrow mesenchymal stem cells enhance fracture healing through the promotion of osteogenesis and angiogenesis in a rat model of nonunion. Stem Cell Res Ther 2020; 11:38. [PMID: 31992369 PMCID: PMC6986095 DOI: 10.1186/s13287-020-1562-9] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/20/2019] [Accepted: 01/12/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND As important players in cell-to-cell communication, exosomes (exo) are believed to play a similar role in promoting fracture healing. This study investigated whether exosomes derived from bone marrow mesenchymal stem cells (BMMSC-Exos) could improve fracture healing of nonunion. METHODS BMMSC-Exos were isolated and transplanted into the fracture site in a rat model of femoral nonunion (Exo group) every week. Moreover, equal volumes of phosphate-buffered saline (PBS) and exosome-depleted conditioned medium (CM-Exo) were injected into the femoral fracture sites of the rats in the control and CM-Exo groups. Bone healing processes were recorded and evaluated by radiographic methods on weeks 8, 14 and 20 after surgery. Osteogenesis and angiogenesis at the fracture sites were evaluated by radiographic and histological methods on postoperative week 20. The expression levels of osteogenesis- or angiogenesis-related genes were evaluated in vitro by western blotting and immunohistochemistry. The ability to internalize exosomes was assessed using the PKH26 assay. Altered proliferation and migration of human umbilical vein endothelial cells (HUVECs) and mouse embryo osteoblast precursor cells (MC3TE-E1s) treated with BMMSC-Exos were determined by utilizing EdU incorporation, immunofluorescence staining, and scratch wound assay. The angiogenesis ability of HUVECs was evaluated through tube formation assays. Finally, to explore the effect of exosomes in osteogenesis via the BMP-2/Smad1/RUNX2 signalling pathway, the BMP-2 inhibitors noggin and LDN193189 were utilized, and their subsequent effects were observed. RESULTS BMMSC-Exos were observed to be spherical with a diameter of approximately 122 nm. CD9, CD63 and CD81 were expressed. Transplantation of BMMSC-Exos obviously enhanced osteogenesis, angiogenesis and bone healing processes in a rat model of femoral nonunion. BMMSC-Exos were taken up by HUVECs and MC3T3-E1 in vitro, and their proliferation and migration were also improved. Finally, experiments with BMP2 inhibitors confirmed that the BMP-2/Smad1/RUNX2 signalling pathway played an important role in the pro-osteogenesis induced by BMMSC-Exos and enhanced fracture healing of nonunion. CONCLUSIONS Our findings suggest that transplantation of BMMSC-Exos exerts a critical effect on the treatment of nonunion by promoting osteogenesis and angiogenesis. This promoting effect might be ascribed to the activation of the BMP-2/Smad1/RUNX2 and the HIF-1α/VEGF signalling pathways.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Spine Surgery, Shandong University Qilu Hospital, Jinan, China
| | - Guangjun Jiao
- Department of Spine Surgery, Shandong University Qilu Hospital, Jinan, China
| | - Shanwu Ren
- Department of Spine Surgery, Shandong University Qilu Hospital, Jinan, China
| | - Xiaoqian Zhang
- Department of Radiology, Shandong University Qilu Hospital, Qingdao, Qingdao, China
| | - Ci Li
- Department of Spine Surgery, Shandong University Qilu Hospital, Jinan, China
| | - Wenliang Wu
- Department of Spine Surgery, Shandong University Qilu Hospital, Jinan, China
| | - Hongliang Wang
- Department of Spine Surgery, Shandong University Qilu Hospital, Jinan, China
| | - Haichun Liu
- Department of Spine Surgery, Shandong University Qilu Hospital, Jinan, China
| | - Hongming Zhou
- Department of Spine Surgery, Shandong University Qilu Hospital, Jinan, China.,Department of Spine Surgery, Linyi Central Hospital, Linyi, China
| | - Yunzhen Chen
- Department of Spine Surgery, Shandong University Qilu Hospital, Jinan, China.
| |
Collapse
|
16
|
Ouyang Z, Tan T, Zhang X, Wan J, Zhou Y, Jiang G, Yang D, Guo X, Liu T. CircRNA hsa_circ_0074834 promotes the osteogenesis-angiogenesis coupling process in bone mesenchymal stem cells (BMSCs) by acting as a ceRNA for miR-942-5p. Cell Death Dis 2019; 10:932. [PMID: 31804461 PMCID: PMC6895238 DOI: 10.1038/s41419-019-2161-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
Bone tissue has a strong ability to repair itself. When treated properly, most fractures will heal well. However, some fractures are difficult to heal. When a fracture does not heal, it is called nonunion. Approximately, 5% of all fracture patients have difficulty healing. Because of the continuous movement of the fracture site, bone nonunion is usually accompanied by pain, which greatly reduces the quality of life of patients. Bone marrow mesenchymal stem cells (BMSCs) play an important role in the process of nonunion. Circular RNAs (circRNAs) are a unique kind of noncoding RNA and represent the latest research hotspot in the RNA field. At present, no studies have reported a role of circRNAs in the development of nonunion. After isolation of BMSCs from patients with nonunion, the expression of circRNAs in these cells was detected by using a circRNA microarray. Alkaline phosphatase and Alizarin red staining were used to detect the regulation of osteogenic differentiation of BMSCs by hsa_circ_0074834. The target gene of hsa_circ_0074834 was detected by RNA pull-down and double-luciferase reporter assay. The ability of hsa_circ_0074834 to regulate the osteogenesis of BMSCs in vivo was tested by heterotopic osteogenesis and single cortical bone defect experiments. The results showed that the expression of hsa_circ_0074834 in BMSCs from patients with nonunion was decreased. Hsa_circ_0074834 acts as a ceRNA to regulate the expression of ZEB1 and VEGF through microRNA-942-5p. Hsa_circ_0074834 can promote osteogenic differentiation of BMSCs and the repair of bone defects. These results suggest that circRNAs may be a key target for the treatment of nonunion.
Collapse
Affiliation(s)
- Zhengxiao Ouyang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Tingting Tan
- Department of Immunology, Xiangya School of Medicine, Central South University, 88 Xiangya Rd., Changsha, Hunan, 410008, P.R. China
| | - Xianghong Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Jia Wan
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Yanling Zhou
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Guangyao Jiang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Daishui Yang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Xiaoning Guo
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Tang Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China.
| |
Collapse
|
17
|
El-Jawhari JJ, Kleftouris G, El-Sherbiny Y, Saleeb H, West RM, Jones E, Giannoudis PV. Defective Proliferation and Osteogenic Potential with Altered Immunoregulatory phenotype of Native Bone marrow-Multipotential Stromal Cells in Atrophic Fracture Non-Union. Sci Rep 2019; 9:17340. [PMID: 31758052 PMCID: PMC6874596 DOI: 10.1038/s41598-019-53927-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/06/2019] [Indexed: 02/08/2023] Open
Abstract
Bone marrow-Multipotential stromal cells (BM-MSCs) are increasingly used to treat complicated fracture healing e.g., non-union. Though, the quality of these autologous cells is not well characterized. We aimed to evaluate bone healing-related capacities of non-union BM-MSCs. Iliac crest-BM was aspirated from long-bone fracture patients with normal healing (U) or non-united (NU). Uncultured (native) CD271highCD45low cells or passage-zero cultured BM-MSCs were analyzed for gene expression levels, and functional assays were conducted using culture-expanded BM-MSCs. Blood samples were analyzed for serum cytokine levels. Uncultured NU-CD271highCD45low cells significantly expressed fewer transcripts of growth factor receptors, EGFR, FGFR1, and FGRF2 than U cells. Significant fewer transcripts of alkaline phosphatase (ALPL), osteocalcin (BGLAP), osteonectin (SPARC) and osteopontin (SPP1) were detected in NU-CD271highCD45low cells. Additionally, immunoregulation-related markers were differentially expressed between NU- and U-CD271highCD45low cells. Interestingly, passage-zero NU BM-MSCs showed low expression of immunosuppressive mediators. However, culture-expanded NU and U BM-MSCs exhibited comparable proliferation, osteogenesis, and immunosuppression. Serum cytokine levels were found similar for NU and U groups. Collectively, native NU-BM-MSCs seemed to have low proliferative and osteogenic capacities; therefore, enhancing their quality should be considered for regenerative therapies. Further research on distorted immunoregulatory molecules expression in BM-MSCs could potentially benefit the prediction of complicated fracture healing.
Collapse
Affiliation(s)
- Jehan J El-Jawhari
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds, UK. .,NIHR Leeds Biomedical Research Centre, Chapel Allerton Hospital, Leeds, UK. .,Clinical pathology department, Mansoura University, Mansoura, Egypt.
| | - George Kleftouris
- Academic Department of Trauma and Orthopaedic, Leeds General Infirmary, School of Medicine, University of Leeds, Leeds, UK
| | - Yasser El-Sherbiny
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds, UK.,Clinical pathology department, Mansoura University, Mansoura, Egypt.,Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Hany Saleeb
- Academic Department of Trauma and Orthopaedic, Leeds General Infirmary, School of Medicine, University of Leeds, Leeds, UK
| | - Robert M West
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Peter V Giannoudis
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Biomedical Research Centre, Chapel Allerton Hospital, Leeds, UK.,Academic Department of Trauma and Orthopaedic, Leeds General Infirmary, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
18
|
Medhat D, Rodríguez CI, Infante A. Immunomodulatory Effects of MSCs in Bone Healing. Int J Mol Sci 2019; 20:ijms20215467. [PMID: 31684035 PMCID: PMC6862454 DOI: 10.3390/ijms20215467] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are capable of differentiating into multilineage cells, thus making them a significant prospect as a cell source for regenerative therapy; however, the differentiation capacity of MSCs into osteoblasts seems to not be the main mechanism responsible for the benefits associated with human mesenchymal stem cells hMSCs when used in cell therapy approaches. The process of bone fracture restoration starts with an instant inflammatory reaction, as the innate immune system responds with cytokines that enhance and activate many cell types, including MSCs, at the site of the injury. In this review, we address the influence of MSCs on the immune system in fracture repair and osteogenesis. This paradigm offers a means of distinguishing target bone diseases to be treated with MSC therapy to enhance bone repair by targeting the crosstalk between MSCs and the immune system.
Collapse
Affiliation(s)
- Dalia Medhat
- Medical Biochemistry Department, National Research Centre, Dokki, Giza 12622, Egypt.
| | - Clara I Rodríguez
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Bizkaia, Spain.
| | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Bizkaia, Spain.
| |
Collapse
|
19
|
Murray CE, Coleman CM. Impact of Diabetes Mellitus on Bone Health. Int J Mol Sci 2019; 20:ijms20194873. [PMID: 31575077 PMCID: PMC6801685 DOI: 10.3390/ijms20194873] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/21/2022] Open
Abstract
Long-term exposure to a diabetic environment leads to changes in bone metabolism and impaired bone micro-architecture through a variety of mechanisms on molecular and structural levels. These changes predispose the bone to an increased fracture risk and impaired osseus healing. In a clinical practice, adequate control of diabetes mellitus is essential for preventing detrimental effects on bone health. Alternative fracture risk assessment tools may be needed to accurately determine fracture risk in patients living with diabetes mellitus. Currently, there is no conclusive model explaining the mechanism of action of diabetes mellitus on bone health, particularly in view of progenitor cells. In this review, the best available literature on the impact of diabetes mellitus on bone health in vitro and in vivo is summarised with an emphasis on future translational research opportunities in this field.
Collapse
Affiliation(s)
- Cliodhna E Murray
- Regenerative Medicine Institute, National University of Ireland, Galway, Biomedical Sciences Building, Dangan, Newcastle Road, Galway City, County Galway, H91W2TY, Ireland.
| | - Cynthia M Coleman
- Regenerative Medicine Institute, National University of Ireland, Galway, Biomedical Sciences Building, Dangan, Newcastle Road, Galway City, County Galway, H91W2TY, Ireland.
| |
Collapse
|
20
|
Mitxitorena I, Infante A, Gener B, Rodríguez CI. Suitability and limitations of mesenchymal stem cells to elucidate human bone illness. World J Stem Cells 2019; 11:578-593. [PMID: 31616536 PMCID: PMC6789184 DOI: 10.4252/wjsc.v11.i9.578] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 07/31/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Functional impairment of mesenchymal stem cells (MSCs), osteoblast progenitor cells, has been proposed to be a pathological mechanism contributing to bone disorders, such as osteoporosis (the most common bone disease) and other rare inherited skeletal dysplasias. Pathological bone loss can be caused not only by an enhanced bone resorption activity but also by hampered osteogenic differentiation of MSCs. The majority of the current treatment options counteract bone loss, and therefore bone fragility by blocking bone resorption. These so-called antiresorptive treatments, in spite of being effective at reducing fracture risk, cannot be administered for extended periods due to security concerns. Therefore, there is a real need to develop osteoanabolic therapies to promote bone formation. Human MSCs emerge as a suitable tool to study the etiology of bone disorders at the cellular level as well as to be used for cell therapy purposes for bone diseases. This review will focus on the most relevant findings using human MSCs as an in vitro cell model to unravel pathological bone mechanisms and the application and outcomes of human MSCs in cell therapy clinical trials for bone disease.
Collapse
Affiliation(s)
- Izaskun Mitxitorena
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo 48903, Bizkaia, Spain
| | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo 48903, Bizkaia, Spain
| | - Blanca Gener
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo 48903, Bizkaia, Spain
- Service of Genetics, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo 48903, Bizkaia, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid 28005, Spain
| | - Clara I Rodríguez
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo 48903, Bizkaia, Spain
| |
Collapse
|
21
|
Long H, Zhu Y, Lin Z, Wan J, Cheng L, Zeng M, Tang Y, Zhao R. miR-381 modulates human bone mesenchymal stromal cells (BMSCs) osteogenesis via suppressing Wnt signaling pathway during atrophic nonunion development. Cell Death Dis 2019; 10:470. [PMID: 31209205 PMCID: PMC6572824 DOI: 10.1038/s41419-019-1693-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 05/19/2019] [Accepted: 05/23/2019] [Indexed: 12/19/2022]
Abstract
The osteogenic differentiation of human bone mesenchymal stromal cells (BMSCs) has been considered as a central issue in fracture healing. Wnt signaling could promote BMSC osteogenic differentiation through inhibiting PPARγ. During atrophic nonunion, Wnt signaling-related factors, WNT5A and FZD3 proteins, were significantly reduced, along with downregulation of Runx2, ALP, and Collagen I and upregulation of PPARγ. Here, we performed a microarray analysis to identify differentially expressed miRNAs in atrophic nonunion tissues that were associated with Wnt signaling through targeting related factors. Of upregulated miRNAs, miR-381 overexpression could significantly inhibit the osteogenic differentiation in primary human BMSCs while increase in PPARγ protein level. Through binding to the 3'UTR of WNT5A and FZD3, miR-381 modulated the osteogenic differentiation via regulating β-catenin nucleus translocation. Moreover, PPARγ, an essential transcription factor inhibiting osteogenic differentiation, could bind to the promoter region of miR-381 to activate its expression. Taken together, PPARγ-induced miR-381 upregulation inhibits the osteogenic differentiation in human BMSCs through miR-381 downstream targets, WNT5A and FZD3, and β-catenin nucleus translocation in Wnt signaling. The in vivo study also proved that inhibition of miR-381 promoted the fracture healing. Our finding may provide a novel direction for atrophic nonunion treatment.
Collapse
Affiliation(s)
- Haitao Long
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yong Zhu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhangyuan Lin
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jun Wan
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Liang Cheng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Min Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yifu Tang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ruibo Zhao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
22
|
Bhattacharjee A, Kuiper JH, Roberts S, Harrison PE, Cassar‐Pullicino VN, Tins B, Bajada S, Richardson JB. Predictors of fracture healing in patients with recalcitrant nonunions treated with autologous culture expanded bone marrow-derived mesenchymal stromal cells. J Orthop Res 2019; 37:1303-1309. [PMID: 30474883 PMCID: PMC6590316 DOI: 10.1002/jor.24184] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/24/2018] [Indexed: 02/04/2023]
Abstract
The study reports the prospective outcome of treating severe recalcitrant fracture nonunion in patients with autologous bone marrow-derived mesenchymal stromal cells (BMSC) from 2003 to 2010 and analyze predictors of union. Autologous BMSC were culture expanded and inserted at nonunion site with or without carriers in addition to surgical stabilization of the fracture. Radiological union was ascertained by musculoskeletal radiologists on plain radiographs and/or CT scans. A logistic regression analysis was performed with cell-expansion parameters (cell numbers, cell doubling time) and known clinical factors (e.g., smoking and diabetes) as independent variables and fracture union as the dependent variable to identify the factors that influence bony healing. An Eq5D index score assessed the effect of treatment on general quality of health. A total of 35 patients (mean age 51+/-13 years) with established nonunion (median 2.9 years, 1-33) and, at least one failed nonunion surgery (median 4,1-14) received treatment. Fracture union was achieved in 21 patients (60%; 95%CI 44-75) at 2.6 years. Multiple penalized logistic regression revealed faster cell doubling time (p = 0.07), absence of diabetes (p = 0.003), less previous surgeries (p = 0.008), and lower age at cell implantation (p = 0.02) were significant predictors for fracture union. A significant increase in Eq5D index (p = 0.01) was noted with a mean rise of the score by 0.34 units (95%CI 0.11-0.58) at 1 year following the study. In summary, the study revealed cell doubling time as a novel in vitro parameter in conjunction with age, multiple surgeries, and diabetes as being significant predictors of the fracture union. © 2018 The Authors. Journal of Orthopaedic Research® Published by Wiley Periodicals, Inc. J Orthop Res 37:1303-1309, 2019.
Collapse
Affiliation(s)
- Atanu Bhattacharjee
- The Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation TrustOswestryUK
| | - Jan H. Kuiper
- The Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation TrustOswestryUK,Institute for Science and Technology in MedicineKeele UniversityKeeleUK
| | - Sally Roberts
- The Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation TrustOswestryUK,Institute for Science and Technology in MedicineKeele UniversityKeeleUK
| | - Paul E. Harrison
- The Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation TrustOswestryUK
| | | | - Bernhard Tins
- The Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation TrustOswestryUK
| | - Stefan Bajada
- The Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation TrustOswestryUK
| | - James B. Richardson
- The Robert Jones and Agnes Hunt Orthopaedic Hospital NHS Foundation TrustOswestryUK,Institute for Science and Technology in MedicineKeele UniversityKeeleUK
| |
Collapse
|
23
|
Kim JP, Seo JB. Editorial Commentary: The Practical Goal of Arthroscopic Osteosynthesis for the Treatment of Unstable Scaphoid Nonunion. Arthroscopy 2018; 34:2819-2820. [PMID: 30286881 DOI: 10.1016/j.arthro.2018.07.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 02/02/2023]
Abstract
Arthroscopic reduction with osteosynthesis using bone grafting has become a successful alternative to open techniques for the treatment of chronically unstable scaphoid nonunions. Several studies have demonstrated that arthroscopic techniques are safe and reproducible in addition to causing less soft tissue damage and providing promising short- and mid-term results. However, these techniques have limitations in restoring normal carpal alignment, especially in patients with unstable scaphoid nonunion and carpal collapse deformities, although this does not affect the recovery of clinical function. Therefore, the practical goal of arthroscopic technique should be kept in mind when treating unstable scaphoid nonunions.
Collapse
|
24
|
Synergistic Effects of Controlled-Released BMP-2 and VEGF from nHAC/PLGAs Scaffold on Osteogenesis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3516463. [PMID: 30345299 PMCID: PMC6174819 DOI: 10.1155/2018/3516463] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 07/12/2018] [Accepted: 09/04/2018] [Indexed: 01/18/2023]
Abstract
Tissue engineering bones take great advantages in massive bone defect repairing; under the induction of growth factors, seed cells differentiate into osteoblasts, and the scaffold materials gradually degrade and are replaced with neogenetic bones, which simulates the actual pathophysiological process of bone regeneration. However, mechanism research is required and further developed to instruct elements selection and optimization. In the present study, we prepared vascular endothelial growth factor/bone morphogenetic protein-2- nanohydroxyapatite/collagen (VEGF/ BMP-2- nHAC/ PLGAs) scaffolds and inoculated mouse MC3T3-E1 preosteoblasts to detect osteogenic indexes and activation of related signaling pathways. The hypothesis is to create a three-dimensional environment that simulates bone defect repairing, and p38 mitogen-activated kinase (p38) inhibitor was applied and osterix shRNA was transferred into mouse MC3T3-E1 preosteoblasts to further investigate the molecular mechanism of crosstalk between BMP-2 and VEGF. Our results demonstrated the following: (1) BMP-2 and VEGF were sustainably released from PLGAs microspheres. (2) nHAC/PLGAs scaffold occupied a three-dimensional porous structure and has excellent physical properties. (3) MC3T3-E1 cells proliferated and differentiated well in the scaffold. (4) Osteogenic differentiation related factors expression of VEGF/BMP-2 loaded scaffold was obviously higher than that of other groups; p38 inhibitor SB203580 decreased the nucleus/cytoplasm ratio of osterix expression. To conclude, the active artificial bone we prepared could provide a favorable growth space for MC3T3-E1 cells, and osteogenesis and maturation reinforced by simultaneous VEGF and BMP-2 treatment may be mainly through the activation of the p38 MAPK pathway to promote nuclear translocation of osterix protein.
Collapse
|
25
|
Perez JR, Kouroupis D, Li DJ, Best TM, Kaplan L, Correa D. Tissue Engineering and Cell-Based Therapies for Fractures and Bone Defects. Front Bioeng Biotechnol 2018; 6:105. [PMID: 30109228 PMCID: PMC6079270 DOI: 10.3389/fbioe.2018.00105] [Citation(s) in RCA: 237] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/09/2018] [Indexed: 12/25/2022] Open
Abstract
Bone fractures and segmental bone defects are a significant source of patient morbidity and place a staggering economic burden on the healthcare system. The annual cost of treating bone defects in the US has been estimated to be $5 billion, while enormous costs are spent on bone grafts for bone injuries, tumors, and other pathologies associated with defective fracture healing. Autologous bone grafts represent the gold standard for the treatment of bone defects. However, they are associated with variable clinical outcomes, postsurgical morbidity, especially at the donor site, and increased surgical costs. In an effort to circumvent these limitations, tissue engineering and cell-based therapies have been proposed as alternatives to induce and promote bone repair. This review focuses on the recent advances in bone tissue engineering (BTE), specifically looking at its role in treating delayed fracture healing (non-unions) and the resulting segmental bone defects. Herein we discuss: (1) the processes of endochondral and intramembranous bone formation; (2) the role of stem cells, looking specifically at mesenchymal (MSC), embryonic (ESC), and induced pluripotent (iPSC) stem cells as viable building blocks to engineer bone implants; (3) the biomaterials used to direct tissue growth, with a focus on ceramic, biodegradable polymers, and composite materials; (4) the growth factors and molecular signals used to induce differentiation of stem cells into the osteoblastic lineage, which ultimately leads to active bone formation; and (5) the mechanical stimulation protocols used to maintain the integrity of the bone repair and their role in successful cell engraftment. Finally, a couple clinical scenarios are presented (non-unions and avascular necrosis—AVN), to illustrate how novel cell-based therapy approaches can be used. A thorough understanding of tissue engineering and cell-based therapies may allow for better incorporation of these potential therapeutic approaches in bone defects allowing for proper bone repair and regeneration.
Collapse
Affiliation(s)
- Jose R Perez
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States.,Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Deborah J Li
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Thomas M Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Lee Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States.,Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
26
|
Shi R, Huang Y, Ma C, Wu C, Tian W. Current advances for bone regeneration based on tissue engineering strategies. Front Med 2018; 13:160-188. [PMID: 30047029 DOI: 10.1007/s11684-018-0629-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/14/2017] [Indexed: 01/07/2023]
Abstract
Bone tissue engineering (BTE) is a rapidly developing strategy for repairing critical-sized bone defects to address the unmet need for bone augmentation and skeletal repair. Effective therapies for bone regeneration primarily require the coordinated combination of innovative scaffolds, seed cells, and biological factors. However, current techniques in bone tissue engineering have not yet reached valid translation into clinical applications because of several limitations, such as weaker osteogenic differentiation, inadequate vascularization of scaffolds, and inefficient growth factor delivery. Therefore, further standardized protocols and innovative measures are required to overcome these shortcomings and facilitate the clinical application of these techniques to enhance bone regeneration. Given the deficiency of comprehensive studies in the development in BTE, our review systematically introduces the new types of biomimetic and bifunctional scaffolds. We describe the cell sources, biology of seed cells, growth factors, vascular development, and the interactions of relevant molecules. Furthermore, we discuss the challenges and perspectives that may propel the direction of future clinical delivery in bone regeneration.
Collapse
Affiliation(s)
- Rui Shi
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Yuelong Huang
- Department of Spine Surgery of Beijing Jishuitan Hospital, The Fourth Clinical Medical College of Peking University, Beijing, 100035, China
| | - Chi Ma
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Chengai Wu
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Wei Tian
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China. .,Department of Spine Surgery of Beijing Jishuitan Hospital, The Fourth Clinical Medical College of Peking University, Beijing, 100035, China.
| |
Collapse
|
27
|
Novel Lipid Signaling Mediators for Mesenchymal Stem Cell Mobilization during Bone Repair. Cell Mol Bioeng 2018; 11:241-253. [PMID: 29983824 DOI: 10.1007/s12195-018-0532-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Introduction Mesenchymal stem and progenitor cells (MSCs), which normally reside in the bone marrow, are critical to bone health and can be recruited to sites of traumatic bone injury, contributing to new bone formation. The ability to control the trafficking of MSCs provides therapeutic potential for improving traumatic bone healing and therapy for genetic bone diseases such as hypophosphatasia. Methods In this study, we explored the sphingosine-1-phosphate (S1P) signaling axis as a means to control the mobilization of MSCs into blood and possibly to recruit MSCs enhancing bone growth. Results Loss of S1P receptor 3 (S1PR3) leads to an increase in circulating CD45-/CD29+/CD90+/Sca1 putative mesenchymal progenitor cells, suggesting that blocking S1PR3 may stimulate MSCs to leave the bone marrow. Antagonism of S1PR3 with the small molecule VPC01091 stimulated acute migration of CD45-/CD29+/CD90+/Sca1+ MSCs into the blood as early as 1.5 hours after treatment. VPC01091 administration also increased ectopic bone formation induced by BMP-2 and significantly increased new bone formation in critically sized rat cranial defects, suggesting that mobilized MSCs may home to injuries to contribute to healing. We also explored the possibility of combining S1P manipulation of endogenous host cell occupancy with exogenous MSC transplantation for potential use in combination therapies. Importantly, reducing niche occupancy of host MSCs with VPC01091 does not impede engraftment of exogenous MSCs. Conclusions Our studies suggest that MSC mobilization through S1PR3 antagonism is a promising strategy for endogenous tissue engineering and improving MSC delivery to treat bone diseases.
Collapse
|
28
|
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
|
29
|
Rupp M, Biehl C, Budak M, Thormann U, Heiss C, Alt V. Diaphyseal long bone nonunions - types, aetiology, economics, and treatment recommendations. INTERNATIONAL ORTHOPAEDICS 2017; 42:247-258. [PMID: 29273837 DOI: 10.1007/s00264-017-3734-5] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 12/12/2017] [Indexed: 12/21/2022]
Abstract
The intention of the current article is to review the epidemiology with related socioeconomic costs, pathophysiology, and treatment options for diaphyseal long bone delayed unions and nonunions. Diaphyseal nonunions in the tibia and in the femur are estimated to occur 4.6-8% after modern intramedullary nailing of closed fractures with an even much higher risk in open fractures. There is a high socioeconomic burden for long bone nonunions mainly driven by indirect costs, such as productivity losses due to long treatment duration. The classic classification of Weber and Cech of the 1970s is based on the underlying biological aspect of the nonunion differentiating between "vital" (hypertrophic) and "avital" (hypo-/atrophic) nonunions, and can still be considered to represent the basis for basic evaluation of nonunions. The "diamond concept" units biomechanical and biological aspects and provides the pre-requisites for successful bone healing in nonunions. For humeral diaphyseal shaft nonunions, excellent results for augmentation plating were reported. In atrophic humeral shaft nonunions, compression plating with stimulation of bone healing by bone grafting or BMPs seem to be the best option. For femoral and tibial diaphyseal shaft fractures, dynamization of the nail is an atraumatic, effective, and cheap surgical possibility to achieve bony consolidation, particularly in delayed nonunions before 24 weeks after initial surgery. In established hypertrophic nonunions in the tibia and femur, biomechanical stability should be addressed by augmentation plating or exchange nailing. Hypotrophic or atrophic nonunions require additional biological stimulation of bone healing for augmentation plating.
Collapse
Affiliation(s)
- Markus Rupp
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Giessen-Marburg GmbH, Campus Giessen, Rudolf-Buchheim-Str. 7, 35385, Giessen, Germany
| | - Christoph Biehl
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Giessen-Marburg GmbH, Campus Giessen, Rudolf-Buchheim-Str. 7, 35385, Giessen, Germany
| | - Matthäus Budak
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Giessen-Marburg GmbH, Campus Giessen, Rudolf-Buchheim-Str. 7, 35385, Giessen, Germany
| | - Ulrich Thormann
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Giessen-Marburg GmbH, Campus Giessen, Rudolf-Buchheim-Str. 7, 35385, Giessen, Germany
| | - Christian Heiss
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Giessen-Marburg GmbH, Campus Giessen, Rudolf-Buchheim-Str. 7, 35385, Giessen, Germany
| | - Volker Alt
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Giessen-Marburg GmbH, Campus Giessen, Rudolf-Buchheim-Str. 7, 35385, Giessen, Germany.
| |
Collapse
|
30
|
A novel cytotherapy device for rapid screening, enriching and combining mesenchymal stem cells into a biomaterial for promoting bone regeneration. Sci Rep 2017; 7:15463. [PMID: 29133959 PMCID: PMC5684202 DOI: 10.1038/s41598-017-15451-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 10/24/2017] [Indexed: 01/26/2023] Open
Abstract
Bone defects are a common challenge in clinic, usually warranting bone grafts. However, current strategies to obtain effective graft materials have many drawbacks. Mesenchymal stem cell (MSC)-based therapy is a promising alternative. We designed an innovative appliance named the stem cell screen-enrich-combine(-biomaterials) circulating system (SECCS). In this study, 42 patients who required bone graft underwent SECCS-based treatment. Their bone marrow samples and beta-tricalcium phosphate (β-TCP) granules were processed in the SECCS for 10-15 minutes, to produce MSC/β-TCP composites. These composites were grafted back into bone defect sites. The results showed 85.53% ± 7.95% autologous MSCs were successfully screened, enriched, and seeded on the β-TCP scaffolds synchronously. The cell viability remained unchanged after SECCS processing. Clinically, all patients obtained satisfactory bone healing. Thus, without in vitro culture, the SECCS can produce bioactive MSC/β-TCP composites for bone regeneration during surgery. The SECCS represents a convenient, rapid, low-cost, and safe method for bone regeneration.
Collapse
|
31
|
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
|
32
|
Cao H, Sun ZB, Zhang L, Qian W, Li CY, Guo XP, Zhang Y. Adenovirus-mediated bone morphogenetic protein-2 promotes osteogenic differentiation in human mesenchymal stem cells in vitro. Exp Ther Med 2017; 14:377-382. [PMID: 28672942 DOI: 10.3892/etm.2017.4482] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 03/17/2017] [Indexed: 01/18/2023] Open
Abstract
Delayed and failed bone union following fracture is a common clinical complication that requires treatment in orthopedics. Cell-based therapies and tissue-engineering approaches are potential therapeutic strategies for bone repair and fracture healing. However, the effect of adenovirus expressing bone morphogenetic protein-2 (Ad-BMP-2) on the osteogenic ability of human mesenchymal stem cells (hMSCs) has remained to be fully elucidated. Therefore, in the present study, hMSCs were transduced using Ad-BMP-2 to assess the effects of its application and to determine whether Ad-BMP-2 promotes the osteogenic differentiation of hMSCs. The purity of the hMSC cultures was assessed using flow cytometric analysis. In order to assess the osteogenic activity, alkaline phosphatase activity (ALP) was measured and to estimate the osteoblastic mineralization and calcification, von Kossa staining for phosphates was performed. Cells positive for Src homology 2 domain were determined to be hMSCs and the presence of CD34 was used to distinguish hematopoietic lineages. Following treatment, the Ad-BMP-2 and control group had significantly increased ALP levels (P<0.05). Compared to the blank group and the group transfected with adenoviral vector containing LacZ, the phosphate deposition in the Ad-BMP-2 group and the positive control group treated with dexamethasone was markedly increased. The results of the present study suggested that Ad-BMP-2 promotes osteogenic differentiation in hMSCs and may have a potential application in treating delayed union and nonunion following bone fracture.
Collapse
Affiliation(s)
- Hong Cao
- Department of Orthopedic Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Zhi-Bo Sun
- Department of Orthopedic Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Lei Zhang
- Department of Orthopedic Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Wei Qian
- Department of Orthopedic Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Chun-Yang Li
- Department of Reproductive Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiao-Peng Guo
- Department of Orthopedic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Ying Zhang
- Department of Reproductive Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
33
|
El-Jawhari JJ, Jones E, Giannoudis PV. The roles of immune cells in bone healing; what we know, do not know and future perspectives. Injury 2016; 47:2399-2406. [PMID: 27809990 DOI: 10.1016/j.injury.2016.10.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Key events occurring during the bone healing include well-orchestrated and complex interactions between immune cells, multipotential stromal cells (MSCs), osteoblasts and osteoclasts. Through three overlapping phases of this physiological process, innate and adaptive immune cells, cytokines and chemokines have a significant role to play. The aim of the escalating immune response is to achieve an osseous healing in the shortest time and with the least complications facilitating the restoration of function. The uninterrupted progression of these biological events in conjunction with a favourable mechanical environment (stable fracture fixation) remains the hallmark of successful fracture healing. When failure occurs, either the biological environment or the mechanical one could have been disrupted. Not infrequently both may be compromised. Consequently, regenerative treatments involving the use of bone autograft, allograft or synthetic matrices supplemented with MSCs are increasingly used. A better understanding of the bone biology and osteoimmunology can help to improve these evolving cell-therapy based strategies. Herein, an up to date status of the role of immune cells during the different phases of bone healing is presented. Additionally, the known and yet to know events about immune cell interactions with MSCs and osteoblasts and osteoclasts and the therapeutic implications are being discussed.
Collapse
Affiliation(s)
- Jehan J El-Jawhari
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St. James Hospital, University of Leeds, UK; NIHR Biomedical Research Unit, Chapel Allerton Hospital, University of Leeds, UK; Clinical Pathology Department, Faculty of Medicine, Mansoura University, Egypt
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St. James Hospital, University of Leeds, UK; NIHR Biomedical Research Unit, Chapel Allerton Hospital, University of Leeds, UK
| | - Peter V Giannoudis
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St. James Hospital, University of Leeds, UK; NIHR Biomedical Research Unit, Chapel Allerton Hospital, University of Leeds, UK.
| |
Collapse
|
34
|
de Seny D, Cobraiville G, Leprince P, Fillet M, Collin C, Mathieu M, Hauzeur JP, Gangji V, Malaise MG. Biomarkers of inflammation and innate immunity in atrophic nonunion fracture. J Transl Med 2016; 14:258. [PMID: 27599571 PMCID: PMC5011805 DOI: 10.1186/s12967-016-1019-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/22/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Nonunion is a failure of healing following a bone fracture. Its physiopathology remains partially unclear and the discovery of new mediators could promote the understanding of bone healing. METHODS Thirty-three atrophic nonunion (NU) patients that failed to demonstrate any radiographic improvement for 6 consecutive months were recruited for providing serum samples. Thirty-five healthy volunteers (HV) served as the control group. Proteomics studies were performed using SELDI-TOF-MS and 2D-DIGE approaches, associated or not with Proteominer® preprocessing, to highlight biomarkers specific to atrophic nonunion pathology. Peak intensities were analyzed by two statistical approaches, a nonparametric Mann-Whitney U tests (univariate approach) and a machine-learning algorithm called extra-trees (multivariate approach). Validation of highlighted biomarkers was performed by alternative approaches such as microfluidic LC-MS/MS, nephelometry, western blotting or ELISA assays. RESULTS From the 35 HV and 33 NU crude serum samples and Proteominer® eluates, 136 spectra were collected by SELDI-TOF-MS using CM10 and IMAC-Cu(2+) ProteinChip arrays, and 665 peaks were integrated for extra-trees multivariate analysis. Accordingly, seven biomarkers and several variants were identified as potential NU biomarkers. Their levels of expression were found to be down- or up-regulated in serum of HV vs NU. These biomarkers are inter-α-trypsin inhibitor H4, hepcidin, S100A8, S100A9, glycated hemoglobin β subunit, PACAP related peptide, complement C3 α-chain. 2D-DIGE experiment allowed to detect 14 biomarkers as being down- or up-regulated in serum of HV vs NU including a cleaved fragment of apolipoprotein A-IV, apolipoprotein E, complement C3 and C6. Several biomarkers such as hepcidin, complement C6, S100A9, apolipoprotein E, complement C3 and C4 were confirmed by an alternative approach as being up-regulated in serum of NU patients compared to HV controls. CONCLUSION Two proteomics approaches were used to identify new biomarkers up- or down-regulated in the nonunion pathology, which are involved in bone turn-over, inflammation, innate immunity, glycation and lipid metabolisms. High expression of hepcidin or S100A8/S100A9 by myeloid cells and the presence of advanced glycation end products and complement factors could be the result of a longstanding inflammatory process. Blocking macrophage activation and/or TLR4 receptor could accelerate healing of fractured bone in at-risk patients.
Collapse
Affiliation(s)
- Dominique de Seny
- Laboratory of Rheumatology, Department of Rheumatology, GIGA Research, University of Liège, Tour GIGA, +2, CHU, 4000, Liège, Belgium.
| | - Gaël Cobraiville
- Laboratory of Rheumatology, Department of Rheumatology, GIGA Research, University of Liège, Tour GIGA, +2, CHU, 4000, Liège, Belgium.,Laboratory for the Analysis of Medicines, Department of Pharmacy, CIRM, University of Liège, 4000, Liège, Belgium
| | - Pierre Leprince
- GIGA-Neurosciences, University of Liège, 4000, Liège, Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines, Department of Pharmacy, CIRM, University of Liège, 4000, Liège, Belgium
| | - Charlotte Collin
- Laboratory of Rheumatology, Department of Rheumatology, GIGA Research, University of Liège, Tour GIGA, +2, CHU, 4000, Liège, Belgium
| | - Myrielle Mathieu
- Laboratory of Bone and Metabolic Biochemistry, Department of Rheumatology, Université Libre de Bruxelles (ULB), 1000, Brussels, Belgium
| | - Jean-Philippe Hauzeur
- Laboratory of Rheumatology, Department of Rheumatology, GIGA Research, University of Liège, Tour GIGA, +2, CHU, 4000, Liège, Belgium
| | - Valérie Gangji
- Laboratory of Bone and Metabolic Biochemistry, Department of Rheumatology, Université Libre de Bruxelles (ULB), 1000, Brussels, Belgium.,Department of Rheumatology and Physical Medicine, Hôpital Erasme, Université Libre de Bruxelles (ULB), 1000, Brussels, Belgium
| | - Michel G Malaise
- Laboratory of Rheumatology, Department of Rheumatology, GIGA Research, University of Liège, Tour GIGA, +2, CHU, 4000, Liège, Belgium
| |
Collapse
|
35
|
Berlier JL, Kharroubi I, Zhang J, Dalla Valle A, Rigutto S, Mathieu M, Gangji V, Rasschaert J. Glucose-Dependent Insulinotropic Peptide Prevents Serum Deprivation-Induced Apoptosis in Human Bone Marrow-Derived Mesenchymal Stem Cells and Osteoblastic Cells. Stem Cell Rev Rep 2016; 11:841-51. [PMID: 26254594 DOI: 10.1007/s12015-015-9616-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human bone marrow-derived mesenchymal stem cells (hBMSC) are able to differentiate into cells of connective tissue lineages, including bone and cartilage. They are therefore considered as a promising tool for the treatment of bone degenerative diseases. One of the major issues in regenerative cell therapy is the biosafety of fetal bovine serum used for cell culture. Therefore, the development of a culture medium devoid of serum but preserving hBMSC viability will be of clinical value. The glucose-dependent insulinotropic peptide (GIP) has an anti-apoptotic action in insulin-producing cells. Interestingly, GIP also exerts beneficial effects on bone turnover by acting on osteoblasts and osteoclasts. We therefore evaluated the ability of GIP to prevent cell death in osteoblastic cells cultured in serum-free conditions. In hBMSC and SaOS-2 cells, activation of the GIP receptor increased intracellular cAMP levels. Serum deprivation induced apoptosis in SaOS-2 and hBMSC that was reduced by 30 and 50 %, respectively, in the presence of GIP. The protective effect of GIP involves activation of the adenylate cyclase pathway and inhibition of caspases 3/7 activation. These findings demonstrate that GIP exerts a protective action against apoptosis in hBMSC and suggest a novel approach to preserve viability of hBMSC cultured in the absence of serum.
Collapse
Affiliation(s)
- J L Berlier
- Laboratory of Bone and Metabolic Biochemistry, Faculty of Medicine, Université libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium
| | - I Kharroubi
- Laboratory of Bone and Metabolic Biochemistry, Faculty of Medicine, Université libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium
| | - J Zhang
- Laboratory of Bone and Metabolic Biochemistry, Faculty of Medicine, Université libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium
| | - A Dalla Valle
- Laboratory of Bone and Metabolic Biochemistry, Faculty of Medicine, Université libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium
| | - S Rigutto
- Laboratory of Bone and Metabolic Biochemistry, Faculty of Medicine, Université libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium
| | - M Mathieu
- Laboratory of Bone and Metabolic Biochemistry, Faculty of Medicine, Université libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium
| | - V Gangji
- Laboratory of Bone and Metabolic Biochemistry, Faculty of Medicine, Université libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium.,Department of Rheumatology and Physical Medicine, Hôpital Erasme, Université libre de Bruxelles, Brussels, Belgium
| | - J Rasschaert
- Laboratory of Bone and Metabolic Biochemistry, Faculty of Medicine, Université libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium.
| |
Collapse
|
36
|
Hao C, Wang Y, Shao L, Liu J, Chen L, Zhao Z. Local Injection of Bone Mesenchymal Stem Cells and Fibrin Glue Promotes the Repair of Bone Atrophic Nonunion In Vivo. Adv Ther 2016; 33:824-33. [PMID: 27098172 DOI: 10.1007/s12325-016-0329-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION This study aimed to evaluate the efficacy of local injection of bone mesenchymal stem cells (BMSCs) and fibrin glue in the treatment of atrophic nonunion in an animal model. METHODS Thirty-six male Lewis rats were randomly assigned into three groups: Group A (control group), Group B (atrophic nonunion group), and Group C (experimental group). All the rats underwent femoral osteotomy of the right hind limb, and stabilized with a custom-designed external fixator. Atrophic nonunion of the rats in Group B and C was induced by cauterization of the periosteum and bone marrow removal, and repaired by injection of fibrin glue and BMSCs-seeded fibrin glue, respectively. The surgically treated femurs were assessed by radiographic and histological analysis, and biomechanical test. RESULTS During the follow-up period, the external fixator maintained correct placement and all the femurs retained normal positioning. Eight weeks postoperatively, atrophic nonunion was detected in Group B, with the presence of fibrous connective tissue in the osteotomy gap. The femurs in Group C demonstrated complete bony bridging of the osteotomy gap, with the formation of plenty of woven bone. CONCLUSION The repair of bone atrophic nonunion can be promoted through local injection of BMSCs and fibrin glue.
Collapse
|
37
|
Das A, Fishero BA, Christophel JJ, Li CJ, Kohli N, Lin Y, Dighe AS, Cui Q. Poly(lactic-co-glycolide) polymer constructs cross-linked with human BMP-6 and VEGF protein significantly enhance rat mandible defect repair. Cell Tissue Res 2015; 364:125-35. [PMID: 26475719 DOI: 10.1007/s00441-015-2301-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 05/21/2015] [Indexed: 11/28/2022]
Abstract
We have previously shown that the combined delivery of mesenchymal stem cells (MSCs), vascular endothelial growth factor (VEGF) and bone morphogenetic protein 6 (BMP-6) induces significantly more bone formation than that induced by the delivery of any single factor or a combination of any two factors. We now determine whether the exogenous addition of VEGF and BMP-6 is sufficient for bone healing when MSCs are not provided. Poly(lactic-co-glycolic acid) (PLAGA) microsphere-based three-dimensional scaffolds (P) were fabricated by thermal sintering of PLAGA microspheres. The scaffolds were chemically cross-linked with 200 ng recombinant human VEGF (P(VEGF)) or BMP-6 (P(BMP-6)) or both (P(VEGF+BMP-6)) by the EDC-NHS-MES method. Release of the proteins from the scaffolds was detected for 21 days in vitro which confirmed their comparable potential to supply the proteins in vivo. The scaffolds were delivered to a critical-sized mandibular defect created in 32 Sprague Dawley rats. Significant bone regeneration was observed only in rats with P(VEGF+BMP-6) scaffolds at weeks 2, 8 and 12 as revealed by micro-computer tomography. Vascular ingrowth was higher in the P(VEGF+BMP-6) group as seen by microfil imaging than in other groups. Trichrome staining revealed that a soft callus formed in P(VEGF), P(BMP-6) and P(VEGF+BMP-6) but not in P. MSCs isolated from rat femurs displayed expression of the bone-specific marker osteocalcin when cultured with P(VEGF), P(BMP-6), or P(VEGF+BMP-6) but not with P. Robust mineralization and increased alkaline phosphatase gene expression were seen in rat MSCs when cultured on P(VEGF+BMP-6) but not on P, P(VEGF), or P(BMP-6). Thus, unlike the delivery of VEGF or BMP-6 alone, the combined delivery of VEGF and BMP-6 to the bone defect significantly enhanced bone repair through the enhancement of angiogenesis and the differentiation of endogenously recruited MSCs into the bone repair site.
Collapse
Affiliation(s)
- Anusuya Das
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Brian A Fishero
- Department of Otolaryngology- Head and Neck Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - J Jared Christophel
- Department of Otolaryngology- Head and Neck Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Ching-Ju Li
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Nikita Kohli
- School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Yong Lin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Abhijit S Dighe
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Quanjun Cui
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
38
|
Li CJ, Madhu V, Balian G, Dighe AS, Cui Q. Cross-Talk Between VEGF and BMP-6 Pathways Accelerates Osteogenic Differentiation of Human Adipose-Derived Stem Cells. J Cell Physiol 2015; 230:2671-82. [PMID: 25753222 DOI: 10.1002/jcp.24983] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 03/03/2015] [Indexed: 12/29/2022]
Abstract
Deficiency in vascular endothelial growth factor (VEGF) or bone morphogenetic proteins (BMPs) results in fracture non-unions. Therefore, it is indispensable to comprehend the combined effect of VEGF and BMPs on the osteogenic differentiation of osteoprogenitor mesenchymal stem cells (MSCs) that are either naturally occurring at the fracture repair site or exogenously added to enhance the bone repair. We found that the combination of VEGF and BMP-6 enhanced COL1A2 expression, which correlated with upregulated expression of osterix, Dlx5, and Msx2 in human adipose-derived stem cells (hADSCs). Cross-talk between VEGF and BMP-6 pathways upregulated activation of p38 mitogen-activated kinase (p38 MAPK) and inhibited activation of protein kinase B (PKB, also known as Akt), whereas phosphorylation of "mothers against decapentaplegic" homologs 1/5/8 (Smads 1/5/8) and extracellular signal-regulated kinases 1 and 2 (ERK 1/2) was not affected. Consistent with these findings, p38 inhibitor SB203580, or siRNA knockdown of osterix, abrogated crosstalk between the VEGF and BMP-6 pathways and significantly reduced the observed upregulation of COL1A2. Nuclear translocation of the phosphorylated form of osterix was also inhibited by SB203580. Although crosstalk between the VEGF-BMP-6 pathways did not show an effect on the extent of mineralization, inhibition of any one of the three components that were upregulated through the cross-talk, i.e., osterix, Dlx5, and p38 activation, led to a complete inhibition of mineralization. Inhibition of PKB/Akt activation, which is attenuated through the cross-talk, significantly enhanced ALP gene expression. These observations imply that crosstalk between the VEGF and BMP-6 signaling pathways enhances osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Ching-Ju Li
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia
| | - Vedavathi Madhu
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia
| | - Gary Balian
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia
| | - Abhijit S Dighe
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia
| | - Quanjun Cui
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
39
|
Herrmann M, Verrier S, Alini M. Strategies to Stimulate Mobilization and Homing of Endogenous Stem and Progenitor Cells for Bone Tissue Repair. Front Bioeng Biotechnol 2015; 3:79. [PMID: 26082926 PMCID: PMC4451737 DOI: 10.3389/fbioe.2015.00079] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/16/2015] [Indexed: 12/17/2022] Open
Abstract
The gold standard for the treatment of critical-size bone defects is autologous or allogenic bone graft. This has several limitations including donor site morbidity and the restricted supply of graft material. Cell-based tissue engineering strategies represent an alternative approach. Mesenchymal stem cells (MSCs) have been considered as a source of osteoprogenitor cells. More recently, focus has been placed on the use of endothelial progenitor cells (EPCs), since vascularization is a critical step in bone healing. Although many of these approaches have demonstrated effectiveness for bone regeneration, cell-based therapies require time consuming and cost-expensive in vitro cell expansion procedures. Accordingly, research is becoming increasingly focused on the homing and stimulation of native cells. The stromal cell-derived factor-1 (SDF-1) - CXCR4 axis has been shown to be critical for the recruitment of MSCs and EPCs. Vascular endothelial growth factor (VEGF) is a key factor in angiogenesis and has been targeted in many studies. Here, we present an overview of the different approaches for delivering homing factors to the defect site by absorption or incorporation to biomaterials, gene therapy, or via genetically manipulated cells. We further review strategies focusing on the stimulation of endogenous cells to support bone repair. Finally, we discuss the major challenges in the treatment of critical-size bone defects and fracture non-unions.
Collapse
Affiliation(s)
| | | | - Mauro Alini
- AO Research Institute Davos , Davos , Switzerland
| |
Collapse
|
40
|
Madhu V, Kilanski A, Reghu N, Dighe AS, Cui Q. Expression of CD105 and CD34 receptors controls BMP-induced in vitro mineralization of mouse adipose-derived stem cells but does not predict their in vivo bone-forming potential. J Orthop Res 2015; 33:625-32. [PMID: 25728702 DOI: 10.1002/jor.22883] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/24/2015] [Indexed: 02/04/2023]
Abstract
Adipose-derived stem cells (ADSCs) can be excellent alternative to bone marrow derived stem cells for enhancing fracture repair since ADSCs can be isolated comparatively in large numbers from discarded lipoaspirates. However, osteogenic potential of ADSCs in vivo is very controversial. We hypothesized that adipose-derived stem cells (ADSCs) that respond maximally to bone morphogenetic proteins (BMPs) in vitro would possess maximum bone-forming potential. Four purified populations of mouse ADSCs: CD105(+) CD34(+), CD105(-) CD34(-), CD105(+) CD34(-) and CD105(-) CD34(+) were obtained using fluorescence-activated cell sorting (FACS) and their BMP-responsiveness was determined in vitro. CD105(+) CD34(-) population showed the strongest response to BMPs in terms of robust increase in mineralization. Expression of CD105 correlated with high BMP-responsive phenotype and larger cell size while expression of CD34 correlated with low BMP-responsive phenotype and smaller cell size. CD105(+) CD34(-) population displayed higher gene expression of Alk1 or Alk6 receptors in comparison with other populations. However, CD105(+) CD34(-) ADSCs failed to induce ectopic bone formation in vivo after they were transplanted into syngeneic mice, indicating that in vitro BMP-responsiveness is not a good indicator to predict in vivo bone forming potential of ADSCs. Therefore greater precautions should be executed during selection of competent ADSCs for bone repair.
Collapse
Affiliation(s)
- Vedavathi Madhu
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, School of Medicine, University of Virginia, Charlottesville, Virginia, 22908
| | | | | | | | | |
Collapse
|
41
|
Kim JP, Seo JB, Yoo JY, Lee JY. Arthroscopic management of chronic unstable scaphoid nonunions: effects on restoration of carpal alignment and recovery of wrist function. Arthroscopy 2015; 31:460-9. [PMID: 25442643 DOI: 10.1016/j.arthro.2014.08.035] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 08/21/2014] [Accepted: 08/28/2014] [Indexed: 02/02/2023]
Abstract
PURPOSE The purpose of this study was to assess the effects of arthroscopically assisted reduction and osteosynthesis on restoration of carpal alignment and recovery of clinical wrist function in patients with unstable scaphoid nonunion. METHODS Thirty-six patients who underwent arthroscopically assisted osteosynthesis with or without bone grafting for unstable scaphoid nonunion between July 2006 and January 2012 were enrolled. The average time from injury to surgery was 51 ± 78.3 months. Radiographic and clinical evaluations were assessed on preoperative and postoperative days, and follow-up evaluation took place at a minimum of 24 months. RESULTS Union was achieved in 86% (31 of 36) of patients at a mean of 11 ± 2.7 weeks. Scaphoid axial length (SAL), lateral intrascaphoid angle (ISA), scapholunate angle (SLA), and reversed carpal height ratio (CHR) was significantly improved after surgery, and those correction ratios averaged 66% ± 46.8%, 74% ± 58.2%, 81% ± 59.8%%, and 94% ± 46%, respectively. The range of wrist motion was unchanged after surgery, but the grip strength improved from 74% ± 22.1% preoperatively to 89% ± 13.7% postoperatively compared with the contralateral side (P = .042). Mean Disabilities of the Arm, Shoulder, and Hand (DASH) and Patient-Related Wrist Evaluation (PRWE) scores improved significantly (P < .001) from 44 and 51 preoperatively to 13 and 23 postoperatively, respectively. The radiological parameters of the scaphoid and carpal alignment in patients who achieved bony union did not correlate with clinical wrist function. CONCLUSIONS Arthroscopic reduction and osteosynthesis of chronic unstable scaphoid nonunion is limited for restoration of normal carpal alignment but has positive effects on the recovery of clinical wrist function. LEVEL OF EVIDENCE Level IV, therapeutic case series.
Collapse
Affiliation(s)
- Jong Pil Kim
- Departments of Orthopedic Surgery and Department of Kinesiology and Medical Science, Dankook University College of Medicine, Cheonan, Republic of Korea.
| | - Joong Bae Seo
- Departments of Orthopedic Surgery and Department of Kinesiology and Medical Science, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Jun Young Yoo
- Departments of Orthopedic Surgery and Department of Kinesiology and Medical Science, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Jee Young Lee
- Department of Diagnostic Radiology, Dankook University College of Medicine, Cheonan, Republic of Korea
| |
Collapse
|
42
|
Madhu V, Li CJ, Dighe AS, Balian G, Cui Q. BMP-non-responsive Sca1+ CD73+ CD44+ mouse bone marrow derived osteoprogenitor cells respond to combination of VEGF and BMP-6 to display enhanced osteoblastic differentiation and ectopic bone formation. PLoS One 2014; 9:e103060. [PMID: 25048464 PMCID: PMC4105618 DOI: 10.1371/journal.pone.0103060] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 06/25/2014] [Indexed: 12/29/2022] Open
Abstract
Clinical trials on fracture repair have challenged the effectiveness of bone morphogenetic proteins (BMPs) but suggest that delivery of mesenchymal stem cells (MSCs) might be beneficial. It has also been reported that BMPs could not increase mineralization in several MSCs populations, which adds ambiguity to the use of BMPs. However, an exogenous supply of MSCs combined with vascular endothelial growth factor (VEGF) and BMPs is reported to synergistically enhance fracture repair in animal models. To elucidate the mechanism of this synergy, we investigated the osteoblastic differentiation of cloned mouse bone marrow derived MSCs (D1 cells) in vitro in response to human recombinant proteins of VEGF, BMPs (-2, -4, -6, -9) and the combination of VEGF with BMP-6 (most potent BMP). We further investigated ectopic bone formation induced by MSCs pre-conditioned with VEGF, BMP-6 or both. No significant increase in mineralization, phosphorylation of Smads 1/5/8 and expression of the ALP, COL1A1 and osterix genes was observed upon addition of VEGF or BMPs alone to the cells in culture. The lack of CD105, Alk1 and Alk6 expression in D1 cells correlated with poor response to BMPs indicating that a greater care in the selection of MSCs is necessary. Interestingly, the combination of VEGF and BMP-6 significantly increased the expression of ALP, COL1A1 and osterix genes and D1 cells pre-conditioned with VEGF and BMP-6 induced greater bone formation in vivo than the non-conditioned control cells or the cells pre-conditioned with either VEGF or BMP-6 alone. This enhanced bone formation by MSCs correlated with higher CADM1 expression and OPG/RANKL ratio in the implants. Thus, combined action of VEGF and BMP on MSCs enhances osteoblastic differentiation of MSCs and increases their bone forming ability, which cannot be achieved through use of BMPs alone. This strategy can be effectively used for bone repair.
Collapse
Affiliation(s)
- Vedavathi Madhu
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ching-Ju Li
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Abhijit S. Dighe
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Gary Balian
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Quanjun Cui
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
43
|
Watson L, Elliman SJ, Coleman CM. From isolation to implantation: a concise review of mesenchymal stem cell therapy in bone fracture repair. Stem Cell Res Ther 2014; 5:51. [PMID: 25099622 PMCID: PMC4055164 DOI: 10.1186/scrt439] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Compromised bone-regenerating capability following a long bone fracture is often the result of reduced host bone marrow (BM) progenitor cell numbers and efficacy. Without surgical intervention, these malunions result in mobility restrictions, deformities, and disability. The clinical application of BM-derived mesenchymal stem cells (MSCs) is a feasible, minimally invasive therapeutic option to treat non-union fractures. This review focuses on novel, newly identified cell surface markers in both the mouse and human enabling the isolation and purification of osteogenic progenitor cells as well as their direct and indirect contributions to fracture repair upon administration. Furthermore, clinical success to date is summarized with commentary on autologous versus allogeneic cell sources and the methodology of cell administration. Given our clinical success to date in combination with recent advances in the identification, isolation, and mechanism of action of MSCs, there is a significant opportunity to develop improved technologies for defining therapeutic MSCs and potential to critically inform future clinical strategies for MSC-based bone regeneration.
Collapse
|
44
|
Rosset P, Deschaseaux F, Layrolle P. Cell therapy for bone repair. Orthop Traumatol Surg Res 2014; 100:S107-12. [PMID: 24411717 DOI: 10.1016/j.otsr.2013.11.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 11/08/2013] [Accepted: 11/15/2013] [Indexed: 02/07/2023]
Abstract
When natural bone repair mechanisms fail, autologous bone grafting is the current standard of care. The osteogenic cells and bone matrix in the graft provide the osteo-inductive and osteo-conductive properties required for successful bone repair. Bone marrow (BM) mesenchymal stem cells (MSCs) can differentiate into osteogenic cells. MSC-based cell therapy holds promise for promoting bone repair. The amount of MSCs available from iliac-crest aspirates is too small to be clinically useful, and either concentration or culture must therefore be used to expand the MSC population. MSCs can be administered alone via percutaneous injection or implanted during open surgery with a biomaterial, usually biphasic hydroxyapatite/β-calcium-triphosphate granules. Encouraging preliminary results have been obtained in patients with delayed healing of long bone fractures or avascular necrosis of the femoral head. Bone tissue engineering involves in vitro MSC culturing on biomaterials to obtain colonisation of the biomaterial and differentiation of the cells. The biomaterial-cell construct is then implanted into the zone to be treated. Few published data are available on bone tissue engineering. Much work remains to be done before determining whether this method is suitable for the routine filling of bone tissue defects. Increasing cell survival and promoting implant vascularisation are major challenges. Improved expertise with culturing techniques, together with the incorporation of regulatory requirements, will open the way to high-quality clinical trials investigating the usefulness of cell therapy as a method for achieving bone repair. Cell therapy avoids the drawbacks of autologous bone grafting, preserving the bone stock and diminishing treatment invasiveness.
Collapse
Affiliation(s)
- P Rosset
- Service de chirurgie orthopédique 2, hôpital Trousseau, Université François-Rabelais de Tours, CHU de Tours, 37044 Tours cedex 09, France; Inserm U957, Laboratoire de physiopathologie de la résorption osseuse et thérapie des tumeurs osseuses primitives (LPRO), Faculté de Médecine, Université de Nantes, Nantes, France.
| | - F Deschaseaux
- StromaLab, UMR CNRS 5273, U1031 Inserm, Établissement Français du Sang Pyrénées-Méditerranée, Université P.-Sabatier, Toulouse, France
| | - P Layrolle
- Inserm U957, Laboratoire de physiopathologie de la résorption osseuse et thérapie des tumeurs osseuses primitives (LPRO), Faculté de Médecine, Université de Nantes, Nantes, France
| |
Collapse
|
45
|
Rőszer T, Józsa T, Kiss-Tóth ED, De Clerck N, Balogh L. Leptin receptor deficient diabetic (db/db) mice are compromised in postnatal bone regeneration. Cell Tissue Res 2013; 356:195-206. [PMID: 24343796 DOI: 10.1007/s00441-013-1768-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 11/04/2013] [Indexed: 12/24/2022]
Abstract
Increased fragility fracture risk with improper healing is a frequent and severe complication of insulin resistance (IR). The mechanisms impairing bone health in IR are still not fully appreciated, which gives importance to studies on bone pathologies in animal models of diabetes. Mice deficient in leptin signaling are widely used models of IR and its comorbidities. Leptin was first recognized as a hormone, regulating appetite and energy balance; however, recent studies have expanded its role showing that leptin is a link between insulin-dependent metabolism and bone homeostasis. In the light of these findings, it is intriguing to consider the role of leptin resistance in bone regeneration. In this study, we show that obese diabetic mice lacking leptin receptor (db/db) are deficient in postnatal regenerative osteogenesis. We apply an ectopic osteogenesis and a fracture healing model, both showing that db/db mice display compromised bone acquisition and regeneration capacity. The underlying mechanisms include delayed periosteal mesenchymatic osteogenesis, premature apoptosis of the cartilage callus and impaired microvascular invasion of the healing tissue. Our study supports the use of the db/db mouse as a model of IR associated bone-healing deficits and can aid further studies of mesenchymatic cell homing and differentiation, microvascular invasion, cartilage to bone transition and callus remodeling in diabetic fracture healing.
Collapse
Affiliation(s)
- Tamás Rőszer
- Department of Cardiovascular Development and Repair, Spanish National Cardiovascular Research Center (Centro Nacional de Investigaciones Cardiovasculares Carlos III), Calle Melchor Fernández Almagro 3, 28029, Madrid, Spain,
| | | | | | | | | |
Collapse
|