1
|
Hedayatzadeh Razavi A, Nafisi N, Velasquez-Hammerle M, Shariyate MJ, Khak M, Mirahmadi A, McNichol M, Rodrogiuez EK, Nazarian A. Advances in computational modeling of cytokine and growth factor dynamics in bone healing: a scoping review. Biomech Model Mechanobiol 2025:10.1007/s10237-025-01938-7. [PMID: 40085288 DOI: 10.1007/s10237-025-01938-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/21/2025] [Indexed: 03/16/2025]
Abstract
Bone healing is a complex process regulated by intricate biological and mechanical factors and spatially varied regions over time. This scoping review synthesizes current computational models that incorporate cytokines and growth factors, examining their role in bone healing. Through a systematic analysis of 71 studies, this review identifies and categorizes the modeling methodologies used, including mathematical, finite element, agent-based, mechanobiological, pharmacobiological, and hybrid approaches. The findings highlight the predominant use of mathematical models while noting a recent shift toward more sophisticated techniques like finite element and agent-based models. Key cytokines and growth factors, such as TGF-β, RANK-RANKL-OPG, and PTH, are repeatedly used, underscoring their essential roles in regulating cellular processes. This review also analyzes parameter selection and validation strategies, identifying gaps in current practices and emphasizing the need for high-quality experimental validation to improve model reliability. Some bibliometric analyses provide insights into citation networks and keyword co-occurrence, illustrating influential studies in the field and central themes. The findings offer a foundation for future research to enhance model accuracy, aiming toward more predictive and clinically relevant models accounting for biology and mechanics in bone healing.
Collapse
Affiliation(s)
- Ahmad Hedayatzadeh Razavi
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN115, Boston, MA, 02215, USA
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Carl J. Shapiro Department of Orthopaedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nazanin Nafisi
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN115, Boston, MA, 02215, USA
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Carl J. Shapiro Department of Orthopaedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Maria Velasquez-Hammerle
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN115, Boston, MA, 02215, USA
- Carl J. Shapiro Department of Orthopaedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mohammad Javad Shariyate
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN115, Boston, MA, 02215, USA
- Carl J. Shapiro Department of Orthopaedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mohammad Khak
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN115, Boston, MA, 02215, USA
- Carl J. Shapiro Department of Orthopaedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alireza Mirahmadi
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN115, Boston, MA, 02215, USA
- Carl J. Shapiro Department of Orthopaedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Megan McNichol
- Knowledge Services, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Edward K Rodrogiuez
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN115, Boston, MA, 02215, USA
- Carl J. Shapiro Department of Orthopaedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ara Nazarian
- Musculoskeletal Translational Innovation Initiative, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN115, Boston, MA, 02215, USA.
- Department of Mechanical Engineering, Boston University, Boston, MA, USA.
- Carl J. Shapiro Department of Orthopaedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Department of Orthopaedic Surgery, Yerevan State Medical University, Yerevan, Armenia.
| |
Collapse
|
2
|
Sharma A, Dutta T, Srivastava A. Underwater Adhesives from Redox-Responsive Polyplexes of Thiolated Polyamide Polyelectrolytes. Chemistry 2024; 30:e202302157. [PMID: 37751057 DOI: 10.1002/chem.202302157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 09/27/2023]
Abstract
We report the fabrication of optically clear underwater adhesives using polyplexes of oppositely charged partially-thiolated polyamide polyelectrolytes (TPEs). The thiol content of the constituent PEs was varied to assess its influence on the adhesive properties of the resulting glues. These catechol-free, redox-responsive TPE-adhesives were formulated in aquo and exhibited high optical transparency and strong adhesion even on submerged or moist surfaces of diverse polar substrates such as glass, aluminium, wood, and bone pieces. The adhesives could be cured under water through oxidative disulphide crosslinking of the constituent TPEs. The polyamide backbone provided multi-site H-bonding interactions with the substrates while the disulphide crosslinking provided the cohesive strength to the glue. Strong adhesion of mammalian bones (load bearing capacity upto 7 kg/cm2 ) was achieved using the adhesive containing 30 mol % thiol residues. Higher pH and use of oxidants such as povidone-iodine solution enhanced the curing rate of the adhesives, and so did the use of Tris buffer instead of Phosphate buffer. The porous architecture of the adhesive and its progressive degradation in aqueous medium over the course of three weeks bode well for diverse biomedical applications where temporary adhesion of tissues is required.
Collapse
Affiliation(s)
- Aashish Sharma
- Department of Chemistry, Indian Institute of Science Education Research, Bhopal, Bhopal By-pass Road, Bhauri, Bhopal, 462066, India
- Current Affiliation: School of Medical and Allied Sciences, G.D. Goenka University, Sohna Road, Gurugram, Haryana, 122103, India
| | - Tanmay Dutta
- Department of Chemistry, Indian Institute of Science Education Research, Bhopal, Bhopal By-pass Road, Bhauri, Bhopal, 462066, India
| | - Aasheesh Srivastava
- Department of Chemistry, Indian Institute of Science Education Research, Bhopal, Bhopal By-pass Road, Bhauri, Bhopal, 462066, India
| |
Collapse
|
3
|
Kendall JJ, Ledoux C, Marques FC, Boaretti D, Schulte FA, Morgan EF, Müller R. An in silico micro-multiphysics agent-based approach for simulating bone regeneration in a mouse femur defect model. Front Bioeng Biotechnol 2023; 11:1289127. [PMID: 38164405 PMCID: PMC10757951 DOI: 10.3389/fbioe.2023.1289127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Bone defects represent a challenging clinical problem as they can lead to non-union. In silico models are well suited to study bone regeneration under varying conditions by linking both cellular and systems scales. This paper presents an in silico micro-multiphysics agent-based (micro-MPA) model for bone regeneration following an osteotomy. The model includes vasculature, bone, and immune cells, as well as their interaction with the local environment. The model was calibrated by time-lapsed micro-computed tomography data of femoral osteotomies in C57Bl/6J mice, and the differences between predicted bone volume fractions and the longitudinal in vivo measurements were quantitatively evaluated using root mean square error (RMSE). The model performed well in simulating bone regeneration across the osteotomy gap, with no difference (5.5% RMSE, p = 0.68) between the in silico and in vivo groups for the 5-week healing period - from the inflammatory phase to the remodelling phase - in the volume spanning the osteotomy gap. Overall, the proposed micro-MPA model was able to simulate the influence of the local mechanical environment on bone regeneration, and both this environment and cytokine concentrations were found to be key factors in promoting bone regeneration. Further, the validated model matched clinical observations that larger gap sizes correlate with worse healing outcomes and ultimately simulated non-union. This model could help design and guide future experimental studies in bone repair, by identifying which are the most critical in vivo experiments to perform.
Collapse
Affiliation(s)
- Jack J. Kendall
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States
| | - Charles Ledoux
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | | | | | - Elise F. Morgan
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Goethals J, Cavazos DR, Denisiuk M, Bray R, Nasr K, Vaidya R. Acute Correction of Femoral Malrotation After Intramedullary Nailing Using an Intraoperative Digital Protractor. Cureus 2023; 15:e37108. [PMID: 37153314 PMCID: PMC10158846 DOI: 10.7759/cureus.37108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2023] [Indexed: 04/07/2023] Open
Abstract
OBJECTIVE The goal of the study is to diagnose and accurately correct malrotation of femur fractures after intramedullary (IM) nailing. MATERIALS AND METHODS An institutional review board (IRB) approved prospective study that was performed at a U.S. level 1 trauma center. After IM nailing of comminuted femur fractures, a computed tomography (CT) scanogram was routinely performed to detect the difference in the postoperative femoral version. Patients with malalignment greater than 15 degrees compared to the contralateral side were informed about the discrepancy and offered to have it acutely corrected. A four-pin technique was used: two Schanz pins were used for measuring angles and two different pins were used to turn and correct the malalignment. The pin in the distal fragment is placed directly under the nail to prevent shortening in comminuted fractures. The nail was unlocked either proximally for retrograde nails or distally for antegrade nails. The Bonesetter Angle application was used as a digital protractor to intraoperatively measure the two reference pins and correct the malrotation. Alternate holes were used for relocking the nail. All patients received a CT scanogram after correction. RESULTS 19/128 patients with comminuted femoral fractures over five years with malrotations between 18 and 47 degrees were included in the study with an average malrotation of 24.7 + 8 degrees. All patients were corrected to an average of 4.0 +/- 2.1 degrees difference, as compared to the contralateral side (range 0-8). No patients required further surgeries to correct malrotation. CONCLUSION Comminuted fractures with malrotation >15 degrees after femoral nailing have an incidence of 15% at our institution. This technique provides an efficient and accurate correction method with the use of an intraoperative digital protractor, avoiding the need for revision IM nailing or osteotomies.
Collapse
|
5
|
Bingol HB, Bender JC, Opsteen JA, Leeuwenburgh SC. Bone adhesive materials: From bench to bedside. Mater Today Bio 2023; 19:100599. [PMID: 37063249 PMCID: PMC10102013 DOI: 10.1016/j.mtbio.2023.100599] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Biodegradable bone adhesives represent a highly sought-after type of biomaterial which would enable replacement of traditional metallic devices for fixation of bone. However, these biomaterials should fulfil an extremely large number of requirements. As a consequence, bone-adhesive biomaterials which meet all of these requirements are not yet commercially available. Therefore, this comprehensive review provides an extensive overview of the development of bone adhesives from a translational perspective. First, the definition, classification, and chemistry of various types of bone adhesives are highlighted to provide a detailed overview of this emerging class of biomaterials. In this review we particularly focused studies which describe the use of materials that are capable of gluing two pieces of bone together within a time frame of minutes to days. Second, this review critically reflects on i) the experimental conditions of commonly employed adhesion tests to assess bone adhesion and ii) the current state-of-the-art regarding their preclinical and clinical applicability.
Collapse
Affiliation(s)
- Hatice B. Bingol
- Department of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
- GATT Technologies BV, Nijmegen, the Netherlands
| | | | | | - Sander C.G. Leeuwenburgh
- Department of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
- Corresponding author.
| |
Collapse
|
6
|
Dixon DT, Gomillion CT. 3D-Printed conductive polymeric scaffolds with direct current electrical stimulation for enhanced bone regeneration. J Biomed Mater Res B Appl Biomater 2023; 111:1351-1364. [PMID: 36825765 DOI: 10.1002/jbm.b.35239] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 12/13/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023]
Abstract
Various methods have been used to treat bone defects caused by genetic disorders, injury, or disease. Yet, there is still great need to develop alternative approaches to repair damaged bone tissue. Bones naturally exhibit piezoelectric potential, or the ability to convert mechanical stresses into electrical impulses. This phenomenon has been utilized clinically to enhance bone regeneration in conjunction with electrical stimulation (ES) therapies; however, oftentimes with critical-sized bone defects, the bioelectric potential at the site of injury is compromised, resulting in less desirable outcomes. In the present study, the potential of a 3D-printed conductive polymer blend to enhance bone formation through restoration of the bioelectrical microenvironment was evaluated. A commercially available 3D printer was used to create circular, thin-film scaffolds consisting of either polylactide (PLA) or a conductive PLA (CPLA) composite. Preosteoblast cells were seeded onto the scaffolds and subjected to direct current ES via a purpose-built cell culture chamber. It was found that CPLA scaffolds had no adverse effects on cell viability, proliferation or differentiation when compared with control scaffolds. The addition of ES, however, resulted in a significant increase in the expression of osteocalcin, a protein indicative of osteoblast maturation, after 14 days of culture. Furthermore, xylenol orange staining also showed the presence of increased mineralized calcium nodules in cultures undergoing stimulation. This study demonstrates the potential for low-cost, conductive scaffolding materials to support cell viability and enhance in vitro mineralization in conjunction with ES.
Collapse
Affiliation(s)
- Damion T Dixon
- School of Environmental, Civil, Agricultural and Mechanical Engineering, College of Engineering, University of Georgia, Athens, Georgia, USA
| | - Cheryl T Gomillion
- School of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
7
|
Jing Z, Liang Z, Yang L, Du W, Yu T, Tang H, Li C, Wei W. Bone formation and bone repair: The roles and crosstalk of osteoinductive signaling pathways. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
8
|
Sibanda V, Anazor F, Relwani J, Dhinsa BS. Outcomes of the Treatment of Fracture Non-union Using Combined Magnetic Field Bone Growth Stimulation: Experiences From a UK Trauma Unit. Cureus 2022; 14:e25100. [PMID: 35733483 PMCID: PMC9205291 DOI: 10.7759/cureus.25100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2022] [Indexed: 11/21/2022] Open
Abstract
Introduction: Fracture non-union is a distressing diagnosis for both patients and clinicians. Several methods have been tried to help promote bone healing. Some of the non-operative strategies include the use of pulsed ultrasound and electrical or magnetic bone stimulators. This study aimed at assessing the outcomes of patients treated with combined magnetic field (CMF) bone stimulators. Methods: All patients with confirmed fracture non-union treated using a CMF bone growth stimulator between May 2019 and December 2021 were included in the study. These were followed up at regular three-month intervals and monitored for signs of clinical and radiological union. The minimum patient follow-up was six months. Our primary outcome measure was union rates following CMF treatment. The secondary outcome measures were time to union and fracture type/configuration in relation to non-union. Results: A total of 29 patients were included. Of the patients, 52% were female. The average age of the patients was 53.42 years (SD: 17.66 years). Four were excluded because their follow-up period was less than six months. Patients were started on CMF bone growth stimulant treatment between four and 27 months from the initial fracture (mean: 11.56 months). The majority of the patients had tibial shaft (21%), distal femur (17%), ankle (10%) and distal humerus (10%) fractures. The overall success rate was 84% (n=21), with a mean time to union of 6.62 months. Conclusion: Bone growth stimulators using combined magnetic fields are a viable treatment option for established fracture non-union. They can result in improved outcomes and can avoid risks and costs associated with surgical options to treat non-union. However, more studies need to be conducted to establish the efficacy of these methods conclusively.
Collapse
|
9
|
Hixon KR, Miller AN. Animal models of impaired long bone healing and tissue engineering- and cell-based in vivo interventions. J Orthop Res 2022; 40:767-778. [PMID: 35072292 DOI: 10.1002/jor.25277] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/05/2021] [Accepted: 01/16/2022] [Indexed: 02/04/2023]
Abstract
Bone healing after injury typically follows a systematic process and occurs spontaneously under appropriate physiological conditions. However, impaired long bone healing is still quite common and may require surgical intervention. Various complications can result in different forms of impaired bone healing including nonunion, critical-size defects, or stress fractures. While a nonunion may occur due to impaired biological signaling and/or mechanical instability, a critical-size defect exhibits extensive bone loss that will not spontaneously heal. Comparatively, a stress fracture occurs from repetitive forces and results in a non-healing crack or break in the bone. Clinical standards of treatment vary between these bone defects due to their pathological differences. The use of appropriate animal models for modeling healing defects is critical to improve current treatment methods and develop novel rescue therapies. This review provides an overview of these clinical bone healing impairments and current animal models available to study the defects in vivo. The techniques used to create these models are compared, along with the outcomes, to clarify limitations and future objectives. Finally, rescue techniques focused on tissue engineering and cell-based therapies currently applied in animal models are specifically discussed to analyze their ability to initiate healing at the defect site, providing information regarding potential future therapies. In summary, this review focuses on the current animal models of nonunion, critical-size defects, and stress fractures, as well as interventions that have been tested in vivo to provide an overview of the clinical potential and future directions for improving bone healing.
Collapse
Affiliation(s)
- Katherine R Hixon
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Thayer School of Engineering, Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Anna N Miller
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Hrkac S, Novak R, Salai G, Grazio S, Vlahovic T, Grgurevic L. Heterotopic ossification vs. fracture healing: Extracellular vesicle cargo proteins shed new light on bone formation. Bone Rep 2022; 16:101177. [PMID: 35252484 PMCID: PMC8892095 DOI: 10.1016/j.bonr.2022.101177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 02/08/2023] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an extremely rare disease in which bone tissue forms in extraskeletal sites, which is known as heterotopic ossification (HO). Extracellular vesicles (EVs) are small phospholipid-enclosed particles released by various cells which have an emerging, but not completely understood role in various (patho)physiological processes. In order to further study the pathophysiology of FOP we conducted a small observational study comparing the proteomic profiles of EV cargo, derived from pooled plasma of four patient groups: FOP patient (N = 1) during active disease phase (flare-up), FOP patients during remission (N = 2), patients after long bone fracture (N = 20) and healthy controls (N = 10). After isolation of EVs – their protein cargo was determined using liquid chromatography / mass spectrometry, after which a functional gene enrichment analysis was performed. Our results show a sizeable difference of the proteomics profiles in which EVs from the bone fracture group show significant activity of integrin interactions, Wnt, VEGF, IGF-1 and PDGF pathways; conversely, FOP patients' EVs indicate that HO occurs via processes of innate immunity and the Ephrin B signaling pathway. We hypothesize that the Ephrin B signaling (expressed in EVs) contributes to HO by aiding in mesenchymal stem cell recruitment and osteogenic differentiation, as well as by contributing to the inflammatory response, including macrophage chemotaxis and activation. This is, to our knowledge, the first published analysis of EV protein cargo in FOP. Proteomics-based analysis of extracellular vesicles’ protein cargo in FOP patients, bone fracture healing and controls. Marked differences in signaling pathways expressed in extracellular vesicles in FOP vs. patients with bone fractures. Ephrin B signaling pathway expressed in extracellular vesicles identified as a likely cogwheel in heterotopic ossification.
Collapse
|
11
|
Kaliya-Perumal AK, Ingham PW. Musculoskeletal regeneration: A zebrafish perspective. Biochimie 2021; 196:171-181. [PMID: 34715269 DOI: 10.1016/j.biochi.2021.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/17/2021] [Accepted: 10/22/2021] [Indexed: 12/18/2022]
Abstract
Musculoskeletal injuries are common in humans. The cascade of cellular and molecular events following such injuries results either in healing with functional recovery or scar formation. While fibrotic scar tissue serves to bridge between injured planes, it undermines functional integrity. Hence, faithful regeneration is the most desired outcome; however, the potential to regenerate is limited in humans. In contrast, various non-mammalian vertebrates have fascinating capabilities of regenerating even an entire appendage following amputation. Among them, zebrafish is an important and accessible laboratory model organism, sharing striking similarities with mammalian embryonic musculoskeletal development. Moreover, clinically relevant muscle and skeletal injury zebrafish models recapitulate mammalian regeneration. Upon muscle injury, quiescent stem cells - known as satellite cells - become activated, proliferate, differentiate and fuse to form new myofibres, while bone fracture results in a phased response involving hematoma formation, inflammation, fibrocartilaginous callus formation, bony callus formation and remodelling. These models are well suited to testing gene- or pharmaco-therapy for the benefit of conditions like muscle tears and fractures. Insights from further studies on whole body part regeneration, a hallmark of the zebrafish model, have the potential to complement regenerative strategies to achieve faster and desired healing following injuries without any scar formation and, in the longer run, drive progress towards the realisation of large-scale regeneration in mammals. Here, we provide an overview of the basic mechanisms of musculoskeletal regeneration, highlight the key features of zebrafish as a regenerative model and outline the relevant studies that have contributed to the advancement of this field.
Collapse
Affiliation(s)
- Arun-Kumar Kaliya-Perumal
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore.
| | - Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore.
| |
Collapse
|
12
|
Chen X, He W, Sun M, Yan Y, Pang Y, Chai G. STING inhibition accelerates the bone healing process while enhancing type H vessel formation. FASEB J 2021; 35:e21964. [PMID: 34694030 DOI: 10.1096/fj.202100069rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022]
Abstract
The stimulator of interferon genes (STING), one of the critical factors of innate immunity, is indicated to be closely related to angiogenesis. This study examined STING's role in angiogenesis and the formation of type H vessels, a specific subtype of bone vessels that regulates bone healing. Different concentrations of 2',3'-cGAMP, and H-151 or C-176 were applied to activate or inhibit STING, respectively. Human umbilical vein endothelial cells were used to examine the effect of STING on angiogenesis in vitro; cell viability, cell migration, and quantitative real-time polymerase chain reactions were performed. Also, the metatarsal experiment was applied as ex vivo proof. Bone fracture or defect mice models were used to examine the effect of STING in vivo; the bone healing process was evaluated by radiography weekly and by μCT on the 14th day after surgery. The formation of type H vessels (CD31hi Emcnhi endothelial cells) and osteogenesis (OCN-positive cells) was assessed using the cryosection and paraffin section. STING activation inhibited angiogenesis both in vitro and ex vivo and slowed down the bone healing process in vivo. Histological analysis showed an increased callus formation, fewer type H vessels, and almost no callus mineralization in the STING activation group compared to the control group. By contrast, H-151 (a hsSTING inhibitor) promoted angiogenesis at a low dose. Moreover, inhibition of mmSTING by C-176 enhanced type H vessels' formation, implying osteogenesis promotion in bone healing (higher bone volume density and more OCN-positive cells). Our data suggested that STING inhibition accelerates the bone healing process while enhancing type H vessel formation.
Collapse
Affiliation(s)
- Xiaojun Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Wenxin He
- Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Mengzhe Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yingjie Yan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yichuan Pang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Gang Chai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Al-Waeli H, Reboucas AP, Mansour A, Morris M, Tamimi F, Nicolau B. Non-steroidal anti-inflammatory drugs and bone healing in animal models-a systematic review and meta-analysis. Syst Rev 2021; 10:201. [PMID: 34238360 PMCID: PMC8268344 DOI: 10.1186/s13643-021-01690-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 04/26/2021] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Non-steroidal anti-inflammatory drugs (NSAID) have excellent anti-inflammatory and analgesic properties and are extensively used to treat post-traumatic or surgical musculoskeletal pain. Although an extensive literature exists on the administration of NSAID on animal bone healing, no systematic review and meta-analysis of animal studies that investigate the effect of NSAID administration on bone fracture healing. Objective of this study is to conduct a systematic review and meta-analysis to estimate the effect of NSAIDs administration on bone healing biomechanical and histomorphometric measurements in different animal models after bone fracture surgery. METHODS We performed a systematic review and meta-analysis of animal studies to estimate the effect of NSAID administration after bone fracture on healing outcomes. We searched eight databases without limiting the search to starting date up to 1 February 2021 for articles on fractured bone healing in animal models in which NSAID were administered. RESULTS Out of 6732 articles screened, 47 were included and 3 common bone healing outcomes were analysed: biomechanical properties (maximum force to break, stiffness, and work-to-failure), micro-computed tomography (μ-CT), and histomorphometric measurements. The studies were generally of low-quality scores because crucial information, especially concerning randomization, blinding, and allocation concealment, was poorly reported. Our results show that the negative effects of NSAID after bone fracture on certain biomechanical properties of the healing bones was not statistically significant in mice compared with other animals, in females compared with males, and in younger compared with older animals. CONCLUSION The findings demonstrated that NSAIDs administration decreased the biomechanical properties of healing bones after fracture surgery in comparison to the control group. Moreover, different effect on certain outcomes was detected among different sites, sex of the animals, and the time of assessment. TRIAL REGISTRATION Protocol published and registered in SYstematic Review Center for Laboratory animal Experimentation (SYRCLE) in 2017, https://www.radboudumc.nl/getmedia/757ec408-7a9e-4635-8233-ae951effea54/Non-Steroidal-Anti-inflammatory-Drugs-and-bone-healing-in-animal-Models-Systematic-Review-and-Meta-Analysis.aspx.
Collapse
Affiliation(s)
- Haider Al-Waeli
- Faculty of Dentistry, Dalhousie University, 5981 University Ave, Halifax, Nova Scotia, B3H 4R2, Canada.
| | - Ana Paula Reboucas
- Faculty of Dentistry, Federal University of Minas Gerais, Minas Gerais, Brazil
- College of Dental Medicine, Qatar University, University Street, Doha, Qatar
| | - Alaa Mansour
- School of Dental Medicine, University at Buffalo, Buffalo, NY, 14214, USA
| | - Martin Morris
- Schulich Library, McGill University, 2001 Avenue McGill College Suite 500, Montréal, QC, H3A 1G1, Canada
| | - Faleh Tamimi
- College of Dental Medicine, Qatar University, University Street, Doha, Qatar
| | - Belinda Nicolau
- Faculty of Dentistry, McGill University, 2001 Avenue McGill College Suite 500, Montréal, QC, H3A 1G1, Canada
| |
Collapse
|
14
|
Luengo-Alonso G, Auñon Martin I, Rodriguez Vega V, Capel Agundez A, Caba Doussoux P. Long term results of lower limb posttraumatic acute bone defects treated with masquelet technique. Injury 2021; 52 Suppl 4:S99-S103. [PMID: 33714549 DOI: 10.1016/j.injury.2021.02.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/10/2021] [Accepted: 02/16/2021] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Several alternatives are now available when treating posttraumatic acute bone defects. Masquelet's technique represents a safe procedure to treat lower limb open fractures associated with significant bone defects and allows surgeons to try to reduce complications incidence. MATERIAL AND METHODS Retrospective study based on patients´ record files and images (2015-2017). Twelve patients suffering acute bone defect, treated using Masquelet technique, were evaluated. A total of ten patients were finally included after exclusion criteria were applied. RESULTS Average time from injury to first stage surgery was 11 days. Free flaps were required in three cases (two latissimus dorsi and one anterolateral thigh flap). Time to second stage surgery was 115 days on average. As bone graft, reaming irrigation-aspiration system was associated to this technique. Consolidation was achieved in all patients but one after an average time of 8.4 months. There was only one case of limb shortening of 20 mm. There was no case of nonunion or infection, and patients could perform daily basic activities independently. VAS mean score was one point one year after final surgery. CONCLUSIONS Masquelet's technique achieves encouraging results when treating lower limb acute bone defects. Using this technique, surgeons could decrease infection incidence in open fractures with severe bone loss. On the other hand, it is a two-stage surgery process, which makes the process longer.
Collapse
|
15
|
Fracture Healing Research-Shift towards In Vitro Modeling? Biomedicines 2021; 9:biomedicines9070748. [PMID: 34203470 PMCID: PMC8301383 DOI: 10.3390/biomedicines9070748] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 01/07/2023] Open
Abstract
Fractures are one of the most frequently occurring traumatic events worldwide. Approximately 10% of fractures lead to bone healing disorders, resulting in strain for affected patients and enormous costs for society. In order to shed light into underlying mechanisms of bone regeneration (habitual or disturbed), and to develop new therapeutic strategies, various in vivo, ex vivo and in vitro models can be applied. Undeniably, in vivo models include the systemic and biological situation. However, transferability towards the human patient along with ethical concerns regarding in vivo models have to be considered. Fostered by enormous technical improvements, such as bioreactors, on-a-chip-technologies and bone tissue engineering, sophisticated in vitro models are of rising interest. These models offer the possibility to use human cells from individual donors, complex cell systems and 3D models, therefore bridging the transferability gap, providing a platform for the introduction of personalized precision medicine and finally sparing animals. Facing diverse processes during fracture healing and thus various scientific opportunities, the reliability of results oftentimes depends on the choice of an appropriate model. Hence, we here focus on categorizing available models with respect to the requirements of the scientific approach.
Collapse
|
16
|
Wallimann A, Magrath W, Thompson K, Moriarty TF, Richards RG, Akdis CA, O’Mahony L, Hernandez CJ. Gut microbial-derived short-chain fatty acids and bone: a potential role in fracture healing. Eur Cell Mater 2021; 41:454-470. [PMID: 33881768 PMCID: PMC9100835 DOI: 10.22203/ecm.v041a29] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Bone healing complications such as delayed healing or non-union affect 5-10 % of patients with a long-bone fracture and lead to reduced quality of life and increased health-care costs. The gut microbiota and the metabolites they produce, mainly short-chain fatty acids (SCFAs), have been shown to impact nearly all organs of the human body including bone. SCFAs show broad activity in positively influencing bone healing outcomes either by acting directly on cell types involved in fracture healing, such as osteoblasts, osteoclasts, chondrocytes and fibroblasts, or indirectly, by shaping an appropriate anti-inflammatory and immune regulatory response. Due to the ability of SCFAs to influence osteoblast and osteoclast differentiation, SCFAs may also affect the integration of orthopaedic implants in bone. In addition, SCFA-derivatives have already been used in a variety of tissue engineering constructs to reduce inflammation and induce bone tissue production. The present review summarises the current knowledge on the role of the gut microbiota, in particular through the action of SCFAs, in the individual stages of bone healing and provides insights into how SCFAs may be utilised in a manner beneficial for fracture healing and surgical reconstruction.
Collapse
Affiliation(s)
- Alexandra Wallimann
- AO Research Institute Davos, Davos, Switzerland,Swiss Institute for Allergy and Asthma Research Davos (SIAF), University of Zurich, Switzerland
| | | | | | | | | | - Cezmi A. Akdis
- Swiss Institute for Allergy and Asthma Research Davos (SIAF), University of Zurich, Switzerland
| | - Liam O’Mahony
- Department of Medicine and School of Microbiology, APC Microbiome Ireland, University College Cork, College Road, Cork, Ireland
| | - Christopher J. Hernandez
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA; Hospital for Special Surgery, New York, NY, USA,Corresponding author: Christopher J. Hernandez (355 Upson Hall, Cornell University, Ithaca, NY 14853 Phone: (607) 255-5129, Fax: (607) 255-1222,
| |
Collapse
|
17
|
Dong L, Song Y, Zhang Y, Zhao W, Wang C, Lin H, Al-Ani MK, Liu W, Xue R, Yang L. Mechanical stretch induces osteogenesis through the alternative activation of macrophages. J Cell Physiol 2021; 236:6376-6390. [PMID: 33634492 DOI: 10.1002/jcp.30312] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/29/2020] [Accepted: 01/20/2021] [Indexed: 02/05/2023]
Abstract
For reconstructive surgeons, critically skeletal damage represents a major challenge. Growing evidence indicate that bone repair is dynamically regulated by the mesenchymal stem cell (MSC)-macrophage interaction. Mechanical strain plays a fundamental role in bone repair and regeneration by influencing MSCs differentiation. Recently, a few findings indicate that macrophages may be mechanically sensitive and their phenotype can be regulated, in part, by mechanical cues. However, how macrophages subjected mechanical stretch influence the osteogenic differentiation of MSCs remain unclear. Thus, the purpose of this study is to explore the effect of macrophages stimulated with mechanical stretch on MSCs osteogenesis. By using a coculture system, we discover that macrophages efficiently induce osteogenic differentiation of MSCs under specific stretch conditions. A synergy mechanism between M2 polarization and YAP/BMP2 axis are identified through molecular and genetic analyses. Macrophages are activated by cyclic stretch and polarized to M2 phenotype that produce anti-inflammatory cytokines such as IL-10 and TGF-β to regulate the local inflammatory microenvironment. Furthermore, mechanical stretch induces YAP activation and nuclear translocation, subsequently regulates downstream BMP2 expression to facilitate MSCs osteogenesis. These findings not only advance our understanding of the complex influence among the mechanical strain, macrophage inflammatory response as well as the osteogenic differentiation of MSCs, but also reveal a control system from mechanical signals to chemical response then to cell behaviors during bone repair and regeneration.
Collapse
Affiliation(s)
- Lili Dong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Yang Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Yajie Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weikang Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunli Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Hungchun Lin
- Columbian College of Art and Science, George Washington University, Washington, District of Columbia, USA
| | - Mohanad Kh Al-Ani
- Department of Microbiology, College of Medicine, Tikrit University, Tikrit, Salahaddin, Iraq
| | - Wanqian Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Ruyue Xue
- Internet Medical and System Applications of National Engineering Laboratory, Zhengzhou, Henan, China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| |
Collapse
|
18
|
Chalikias S, Papaioannou N, Koundis G, Pappa E, Galanos A, Anastassopoulos G, Sarris IN, Panteliou S, Chronopoulos E, Dontas IA. Evaluation of Femoral Bone Fracture Healing in Rats by the Modal Damping Factor and Its Correlation With Peripheral Quantitative Computed Tomography. Cureus 2021; 13:e13342. [PMID: 33754085 PMCID: PMC7971724 DOI: 10.7759/cureus.13342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Introduction Monitoring the progress of fracture healing is essential in order to establish the appropriate timing that ensures adequate bone strength for weight-bearing. In the present experimental study on a rat model of femoral fracture healing, the measurement of bone density and strength by peripheral quantitative computerized tomography (pQCT) was correlated with the modal damping factor (MDF) method. Methods Four groups of 12 male six-month-old Wistar rats each were anesthetized and submitted to baseline femoral pQCT and MDF scanning, followed by aseptic midshaft osteotomy of the right femur which was fixed by a locking intramedullary nail technique. The animals were left to recover and re-scanned following euthanasia of each group after six, eight, 10, and 12 weeks, respectively. The parameters measured by the pQCT method were total bone mineral density (BMD) and polar strength strain index (SSIp). Results Fracture healing progressed over time and at 12 weeks post-osteotomy there was no statistically significant difference between the osteotomized right and the control left femurs regarding MDF, BMD, and SSIp measurements. The highest correlations for the osteotomized femurs were observed between MDF and BMD (r = -0.647, P = 0.043), and between MDF and SSIp (r = -0.350, P = 0.321), at 10 weeks postoperatively. The high to moderate correlations between MDF and BMD, and between MDF and SSIp respectively, support the validity of MDF in assessing fracture healing. Conclusions Based on our findings in this fracture healing animal model, the results from the MDF method are reliable and correlate highly with the total BMD and moderately with the SSI polar values obtained by the pQCT method of bone quality measurement. Further studies are needed which may additionally support that the MDF method can be an attractive portable alternative to monitor fracture healing in the community.
Collapse
Affiliation(s)
- Stavros Chalikias
- Department of Orthopedics, Golden Jubilee National Hospital, Glasgow, GBR
| | - Nikolaos Papaioannou
- Laboratory for Research of the Musculoskeletal System, KAT General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, GRC
| | - George Koundis
- 5th Department of Orthopedics, KAT General Hospital, Athens, GRC
| | - Eleni Pappa
- 5th Department of Orthopedics, KAT General Hospital, Athens, GRC
| | - Antonios Galanos
- Laboratory for Research of the Musculoskeletal System, KAT General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, GRC
| | | | - Ioannis N Sarris
- Department of Mechanical Engineering and Aeronautics, University of Patras, Patras, GRC
| | - Sofia Panteliou
- Department of Mechanical Engineering and Aeronautics, University of Patras, Patras, GRC
| | - Efstathios Chronopoulos
- Laboratory for Research of the Musculoskeletal System, KAT General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, GRC.,2nd Department of Orthopedics, Konstantopouleio General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, GRC
| | - Ismene A Dontas
- Laboratory for Research of the Musculoskeletal System, KAT General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, GRC
| |
Collapse
|
19
|
Moore ER, Mathews OA, Yao Y, Yang Y. Prx1-expressing cells contributing to fracture repair require primary cilia for complete healing in mice. Bone 2021; 143:115738. [PMID: 33188955 PMCID: PMC7769995 DOI: 10.1016/j.bone.2020.115738] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/25/2020] [Accepted: 11/07/2020] [Indexed: 02/09/2023]
Abstract
Bone is a dynamic organ that is continuously modified during development, load-induced adaptation, and fracture repair. Understanding the cellular and molecular mechanisms for natural fracture healing can lead to therapeutics that enhance the quality of newly formed tissue, advance the rate of healing, or replace the need for invasive surgical procedures. Prx1-expressing cells in the periosteum are thought to supply the majority of osteoblasts and chondrocytes in the fracture callus, but the exact mechanisms for this behavior are unknown. The primary cilium is a sensory organelle that is known to mediate several signaling pathways involved in fracture healing and required for Prx1-expressing cells to contribute to juvenile bone development and adult load-induced bone formation. We therefore investigated the role of Prx1-expressing cell primary cilia in fracture repair by developing a mouse model that enabled us to simultaneously track Prx1 lineage cell fate and disrupt Prx1-expressing cell primary cilia in vivo. The cilium KO mice exhibited abnormally large calluses with significantly decreased bone formation and persistent cartilage nodules. Analysis of mRNA expression in the early soft callus revealed downregulation of osteogenesis, Hh signaling, and Wnt signaling, and upregulation of chondrogenesis and angiogenesis. The mutant mice also exhibited decreased Osx and Periostin but increased αSMA and PECAM-1 protein expression in the hard callus. We further used a Gli1LacZ reporter and found that Hh signaling was significantly upregulated in the mutant callus at later stages of healing. Interestingly, altered protein expression and Hh signaling did not correlate with labeled Prx1-lineage cells, suggesting loss of cilia altered Hh signaling non-autonomously. Overall, cilium KO mice demonstrated severely delayed and incomplete fracture healing, and our findings suggest Prx1-expressing cell primary cilia are necessary to tune Hh signaling for proper fracture repair.
Collapse
Affiliation(s)
| | - O Amandhi Mathews
- Harvard School of Dental Medicine, Boston, MA, USA; University of Dallas, Irving, TX, USA
| | - Yichen Yao
- Harvard School of Dental Medicine, Boston, MA, USA; Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingzi Yang
- Harvard School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
20
|
Shen L, Xiao Y, Xie H, Zhao H, Luo T, Liu L, Pan X. A naturally derived small molecule NDSM253 inhibits IKK1 to suppress inflammation response and promote bone healing after fracture. Am J Transl Res 2021; 13:24-37. [PMID: 33527006 PMCID: PMC7847520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/04/2020] [Indexed: 06/12/2023]
Abstract
Bone fracture induces an acute inflammatory response in the resident and peripheral monocyte/macrophage cells. Excessive amounts of proinflammatory cytokines can cause severe tissue damage and inhibit bone healing. The proinflammatory cytokine genes are mainly controlled by TLR4/NF-κB (Toll-like receptor 4/Nuclear factor κB). Thus, targeting the molecules in this signaling pathway to decrease the expression of proinflammatory cytokines is an effective strategy to inhibit the inflammatory response. Herein, we identified a naturally derived small molecule NDSM253 that specifically inhibited IKKα (Inhibitor of NF-κB kinase subunit-alpha), a critical component of TLR4/NF-κB signaling. Biochemically, NDMS253 decreased phosphorylation of IκB (Inhibitor of NF-κB), thereby increasing the binding of IκB-NF-κB and suppressing the proinflammatory cytokine gene expression. NDMS253 showed a much stronger inhibitory effect on proinflammatory cytokine gene expression than did the known IKK inhibitors, including ACHP (2-Amino-6-[2-(cyclopropylmethoxy)-6-hydroxyphenyl]-4-(4-piperidinyl)-3-pyridinecarbonitrile), IKK16, and Amlexanox. Administration of these IKK inhibitors in a mouse femoral fracture model showed that NDSM253 suppressed proinflammatory cytokine genes, thereby promoting bone healing, while the other three IKK inhibitors showed a weaker improvement of both bone healing and circulating proinflammatory cytokines. Collectively, our data suggested that NDSM253 might be an effective inhibitor of IKKα that could inhibit inflammatory cytokine action in bone injury.
Collapse
Affiliation(s)
- Liqi Shen
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Yun Xiao
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Hui Xie
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Hongbin Zhao
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Tao Luo
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Lin Liu
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| | - Xuekun Pan
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province Kunming, Yunnan, China
| |
Collapse
|
21
|
Li Y, Hoffman MD, Benoit DSW. Matrix metalloproteinase (MMP)-degradable tissue engineered periosteum coordinates allograft healing via early stage recruitment and support of host neurovasculature. Biomaterials 2021; 268:120535. [PMID: 33271450 PMCID: PMC8110201 DOI: 10.1016/j.biomaterials.2020.120535] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/17/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022]
Abstract
Despite serving as the clinical "gold standard" treatment for critical size bone defects, decellularized allografts suffer from long-term failure rates of ~60% due to the absence of the periosteum. Stem and osteoprogenitor cells within the periosteum orchestrate autograft healing through host cell recruitment, which initiates the regenerative process. To emulate periosteum-mediated healing, tissue engineering approaches have been utilized with mixed outcomes. While vascularization has been widely established as critical for bone regeneration, innervation was recently identified to be spatiotemporally regulated together with vascularization and similarly indispensable to bone healing. Notwithstanding, there are no known approaches that have focused on periosteal matrix cues to coordinate host vessel and/or axon recruitment. Here, we investigated the influence of hydrogel degradation mechanism, i.e. hydrolytic or enzymatic (cell-dictated), on tissue engineered periosteum (TEP)-modified allograft healing, especially host vessel/nerve recruitment and integration. Matrix metalloproteinase (MMP)-degradable hydrogels supported endothelial cell migration from encapsulated spheroids whereas no migration was observed in hydrolytically degradable hydrogels in vitro, which correlated with increased neurovascularization in vivo. Specifically, ~2.45 and 1.84-fold, and ~3.48 and 2.58-fold greater vessel and nerve densities with high levels of vessel and nerve co-localization was observed using MMP degradable TEP (MMP-TEP) -modified allografts versus unmodified and hydrolytically degradable TEP (Hydro-TEP)-modified allografts, respectively, at 3 weeks post-surgery. MMP-TEP-modified allografts exhibited greater longitudinal graft-localized vascularization and endochondral ossification, along with 4-fold and 2-fold greater maximum torques versus unmodified and Hydro-TEP-modified allografts after 9 weeks, respectively, which was comparable to that of autografts. In summary, our results demonstrated that the MMP-TEP coordinated allograft healing via early stage recruitment and support of host neurovasculature.
Collapse
Affiliation(s)
- Yiming Li
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| | - Michael D Hoffman
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA; Materials Science Program, University of Rochester, Rochester, NY, USA; Department of Chemical Engineering, University of Rochester, Rochester, NY, USA; Department of Biomedical Genetics and Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
22
|
|
23
|
Kegelman CD, Nijsure MP, Moharrer Y, Pearson HB, Dawahare JH, Jordan KM, Qin L, Boerckel JD. YAP and TAZ Promote Periosteal Osteoblast Precursor Expansion and Differentiation for Fracture Repair. J Bone Miner Res 2021; 36:143-157. [PMID: 32835424 PMCID: PMC7988482 DOI: 10.1002/jbmr.4166] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/10/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022]
Abstract
In response to bone fracture, periosteal progenitor cells proliferate, expand, and differentiate to form cartilage and bone in the fracture callus. These cellular functions require the coordinated activation of multiple transcriptional programs, and the transcriptional regulators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) regulate osteochondroprogenitor activation during endochondral bone development. However, recent observations raise important distinctions between the signaling mechanisms used to control bone morphogenesis and repair. Here, we tested the hypothesis that YAP and TAZ regulate osteochondroprogenitor activation during endochondral bone fracture healing in mice. Constitutive YAP and/or TAZ deletion from Osterix-expressing cells impaired both cartilage callus formation and subsequent mineralization. However, this could be explained either by direct defects in osteochondroprogenitor differentiation after fracture or by developmental deficiencies in the progenitor cell pool before fracture. Consistent with the second possibility, we found that developmental YAP/TAZ deletion produced long bones with impaired periosteal thickness and cellularity. Therefore, to remove the contributions of developmental history, we next generated adult onset-inducible knockout mice (using Osx-CretetOff ) in which YAP and TAZ were deleted before fracture but after normal development. Adult onset-induced YAP/TAZ deletion had no effect on cartilaginous callus formation but impaired bone formation at 14 days post-fracture (dpf). Earlier, at 4 dpf, adult onset-induced YAP/TAZ deletion impaired the proliferation and expansion of osteoblast precursor cells located in the shoulder of the callus. Further, activated periosteal cells isolated from this region at 4 dpf exhibited impaired osteogenic differentiation in vitro upon YAP/TAZ deletion. Finally, confirming the effects on osteoblast function in vivo, adult onset-induced YAP/TAZ deletion impaired bone formation in the callus shoulder at 7 dpf before the initiation of endochondral ossification. Together, these data show that YAP and TAZ promote the expansion and differentiation of periosteal osteoblast precursors to accelerate bone fracture healing. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Christopher D Kegelman
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Yasaman Moharrer
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Hope B Pearson
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - James H Dawahare
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Kelsey M Jordan
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Sabouni K, Ozturk Y, Kacar E, Mutlu HS, Solakoglu S, Kose GT, Kok FN, Kazmanli MK, Urgen KM, Onder S. Assessment of bone healing using (Ti,Mg)N thin film coated plates and screws: Rabbit femur model. J Biomed Mater Res B Appl Biomater 2020; 109:227-237. [PMID: 32770599 DOI: 10.1002/jbm.b.34694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 07/03/2020] [Accepted: 07/09/2020] [Indexed: 11/06/2022]
Abstract
Magnesium (Mg) based implants such as plates and screws are often preferred to treat bone defects because of the positive effects of magnesium in bone growth and healing. Their low corrosion resistance, however, leads to fast degradation and consequently failure before healing was completed. Previously, we developed Mg doped titanium nitrate (TiN) thin film coatings to address these limitations and demonstrated that <10 at% Mg doping led to enhanced mineralization in vitro. In the present study, in vivo performance of (Ti,Mg)N coated Ti6Al4V based plates and screws were studied in the rabbit model. Bone fractures were formed on femurs of 16 rabbits and then fixed with either (Ti,Mg)N coated (n = 8) or standard TiN coated (n = 8) plates and screws. X-ray imaging and μCT analyses showed enhanced bone regeneration on fracture sites fixed with (Ti,Mg)N coated plates in comparison with the Mg free ones. Bone mineral density, bone volume, and callus volume were also found to be 11.4, 23.4, and 42.8% higher, respectively, in accordance with μCT results. Furthermore, while TiN coatings promoted only primary bone regeneration, (Ti,Mg)N led to secondary bone regeneration in 6 weeks. These results indicated that Mg presence in the coatings accelerated bone regeneration in the fracture site. (Ti,Mg)N coating can be used as a practical method to increase the efficiency of existing bone fixation devices of varying geometry.
Collapse
Affiliation(s)
- Kenda Sabouni
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | - Yetkin Ozturk
- Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul, Turkey
| | - Erkan Kacar
- Department of Metallurgical and Materials Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Hasan Serdar Mutlu
- Department of Histology and Embryology, Istanbul University, Istanbul, Turkey
| | - Seyhun Solakoglu
- Department of Histology and Embryology, Istanbul University, Istanbul, Turkey
| | - Gamze Torun Kose
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | - Fatma Nese Kok
- Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul, Turkey
| | - Muhammet Kursat Kazmanli
- Department of Metallurgical and Materials Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Kamil Mustafa Urgen
- Department of Metallurgical and Materials Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Sakip Onder
- Department of Biomedical Engineering, Yildiz Technical University, Istanbul, Turkey
| |
Collapse
|
25
|
Park YH, Cho HW, Choi GW, Kim HJ. Necessity of Interfragmentary Lag Screws in Precontoured Lateral Locking Plate Fixation for Supination-External Rotation Lateral Malleolar Fractures. Foot Ankle Int 2020; 41:818-826. [PMID: 32389050 DOI: 10.1177/1071100720917645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Interfragmentary lag screws, protected by a plate, have been applied for many years in the treatment of supination-external rotation (SER) ankle fractures. Recently, similar biomechanical stability was found between fixation completed with a plate and lag screw and a plate alone. The aim of this study was to determine whether interfragmentary lag screws are necessary during precontoured lateral locking plate fixation for SER lateral malleolar fractures. METHODS A prospective randomized controlled trial of 76 patients with unilateral Lauge-Hansen SER lateral malleolar fractures was conducted. The patients were randomly treated either with or without the use of interfragmentary lag screws with precontoured lateral locking plate fixation. Clinical outcomes were assessed using the Olerud-Molander Ankle Score and a visual analog scale for pain. Radiologic outcomes were assessed based on the Kellgren and Lawrence scale score, incongruity of the ankle joint, and type of fracture healing. Sixty-nine patients completed 12 months of follow-up. RESULTS There was no significant difference between the 2 groups with regard to clinical outcomes at 3 and 12 months after surgery and radiologic outcomes at 12 months after surgery. All patients in both groups achieved primary bone healing. CONCLUSION The results of this study suggest that with precontoured lateral locking plate fixation, the use of interfragmentary lag screw is not essential in the treatment SER lateral malleolar fractures. LEVEL OF EVIDENCE Level I, prospective randomized study.
Collapse
Affiliation(s)
- Young Hwan Park
- Department of Orthopaedic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Hyun Woo Cho
- Department of Orthopaedic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Gi Won Choi
- Department of Orthopaedic Surgery, Korea University Ansan Hospital, Seoul, Korea
| | - Hak Jun Kim
- Department of Orthopaedic Surgery, Korea University Guro Hospital, Seoul, Korea
| |
Collapse
|
26
|
Martineau C, Kaufmann M, Arabian A, Jones G, St-Arnaud R. Preclinical safety and efficacy of 24R,25-dihydroxyvitamin D 3 or lactosylceramide treatment to enhance fracture repair. J Orthop Translat 2020; 23:77-88. [PMID: 32518749 PMCID: PMC7270532 DOI: 10.1016/j.jot.2020.03.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/05/2020] [Accepted: 03/25/2020] [Indexed: 01/22/2023] Open
Abstract
Background/Objective Cyp24a1-null mice deficient in 24,25(OH)2D3 display impaired callus formation during the endochondral phase of bone fracture repair. The 24,25(OH)2D3 metabolite acted by binding to the TLC domain containing 3B isoform 2 (TLCD3B2, previously named FAM57B2) effector protein, which then synthesizes lactosylceramide (LacCer). Treatment with 24,25(OH)2D3 or LacCer restored callus size and mechanical properties in Cyp24a1-null mice. Methods To assess the safety of these molecules and test their efficacy for bone healing in wild-type, non-genetically modified mice, we treated 12-week-old, osteotomized C57BL/6 female mice with each compound for up to 21 days post-osteotomy. Control cohorts were injected with vehicle. Results Neither compound was found to exhibit any nephro- nor hepato-toxicity. Calcemia remained stable throughout the experiment and was unaffected by either treatment. Supplementation with 24,25(OH)2D3 increased circulating levels of this metabolite about 8-fold, decreased 1,25(OH)2D3 levels, and significantly increased circulating 1,24,25(OH)3D3 levels, suggesting 1?-hydroxylation of 24,25(OH)2D3. TLCD3B2 was found to be expressed in fracture callus at the surface of unmineralized or pre-mineralized cartilage on day 10 and day 12 post-osteotomy and to progressively recede to become undetectable by day 18. Treatment with 24,25(OH)2D3 or LacCer reduced the number of TLCD3B2-positive cells. Both treatments also significantly increased stiffness and elastic modulus of the healing bone callus. Conclusion Exogenous administration of 24,25(OH)2D3 or LacCer improved the biomechanical properties of repaired bones in wild-type animals without affecting circulating calcium levels or other blood parameters, demonstrating preclinical safety and efficacy. Translational potential Our data suggest the use of 24R,25-dihydroxyvitamin D3 or lactosylceramide for ameliorating fracture healing in clinical practice.
Collapse
Affiliation(s)
- Corine Martineau
- Research Centre, Shriners Hospitals for Children – Canada, Montreal, Quebec, H4A 0A9, Canada
| | - Martin Kaufmann
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
- Department of Surgery, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Alice Arabian
- Research Centre, Shriners Hospitals for Children – Canada, Montreal, Quebec, H4A 0A9, Canada
| | - Glenville Jones
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - René St-Arnaud
- Research Centre, Shriners Hospitals for Children – Canada, Montreal, Quebec, H4A 0A9, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Department of Surgery, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Department of Medicine, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Research Institute of the McGill University Health Centre, Montreal, Quebec, H3H 2R9, Canada
- Corresponding author. Research Centre, Shriners Hospitals for Children – Canada, 1003 Decarie Boulevard, Montreal, Quebec, H4A 0A9, Canada.
| |
Collapse
|
27
|
Obeid BA. Implants and grafts used in fractures for early healing. JOURNAL OF ORTHOPAEDICS AND SPINE 2020. [DOI: 10.4103/joas.joas_45_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
28
|
Buettmann EG, McKenzie JA, Migotsky N, Sykes DA, Hu P, Yoneda S, Silva MJ. VEGFA From Early Osteoblast Lineage Cells (Osterix+) Is Required in Mice for Fracture Healing. J Bone Miner Res 2019; 34:1690-1706. [PMID: 31081125 PMCID: PMC6744295 DOI: 10.1002/jbmr.3755] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/23/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022]
Abstract
Bone formation via intramembranous and endochondral ossification is necessary for successful healing after a wide range of bone injuries. The pleiotropic cytokine, vascular endothelial growth factor A (VEGFA) has been shown, via nonspecific pharmacologic inhibition, to be indispensable for angiogenesis and ossification following bone fracture and cortical defect repair. However, the importance of VEGFA expression by different cell types during bone healing is not well understood. We sought to determine the role of VEGFA from different osteoblast cell subsets following clinically relevant models of bone fracture and cortical defect. Ubiquitin C (UBC), Osterix (Osx), or Dentin matrix protein 1 (Dmp1) Cre-ERT2 mice (male and female) containing floxed VEGFA alleles (VEGFAfl/fl ) were either given a femur full fracture, ulna stress fracture, or tibia cortical defect at 12 weeks of age. All mice received tamoxifen continuously starting 2 weeks before bone injury and throughout healing. UBC Cre-ERT2 VEGFAfl/fl (UBC cKO) mice, which were used to mimic nonspecific inhibition, had minimal bone formation and impaired angiogenesis across all bone injury models. UBC cKO mice also exhibited impaired periosteal cell proliferation during full fracture, but not stress fracture repair. Osx Cre-ERT2 VEGFAfl/fl (Osx cKO) mice, but not Dmp1 Cre-ERT2 VEGFAfl/fl (Dmp1 cKO) mice, showed impaired periosteal bone formation and angiogenesis in models of full fracture and stress fracture. Neither Osx cKO nor Dmp1 cKO mice demonstrated significant impairments in intramedullary bone formation and angiogenesis following cortical defect. These data suggest that VEGFA from early osteolineage cells (Osx+), but not mature osteoblasts/osteocytes (Dmp1+), is critical at the time of bone injury for rapid periosteal angiogenesis and woven bone formation during fracture repair. Whereas VEGFA from another cell source, not from the osteoblast cell lineage, is necessary at the time of injury for maximum cortical defect intramedullary angiogenesis and osteogenesis. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicole Migotsky
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - David Aw Sykes
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Pei Hu
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Susumu Yoneda
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
29
|
Mazini L, Rochette L, Amine M, Malka G. Regenerative Capacity of Adipose Derived Stem Cells (ADSCs), Comparison with Mesenchymal Stem Cells (MSCs). Int J Mol Sci 2019; 20:ijms20102523. [PMID: 31121953 PMCID: PMC6566837 DOI: 10.3390/ijms20102523] [Citation(s) in RCA: 262] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue is now on the top one of stem cell sources regarding its accessibility, abundance, and less painful collection procedure when compared to other sources. The adipose derived stem cells (ADSCs) that it contains can be maintained and expanded in culture for long periods of time without losing their differentiation capacity, leading to large cell quantities being increasingly used in cell therapy purposes. Many reports showed that ADSCs-based cell therapy products demonstrated optimal efficacy and efficiency in some clinical indications for both autologous and allogeneic purposes, hence becoming considered as potential tools for replacing, repairing, and regenerating dead or damaged cells. In this review, we analyzed the therapeutic advancement of ADSCs in comparison to bone marrow (BM) and umbilical cord (UC)-mesenchymal stem cells (MSCs) and designed the specific requirements to their best clinical practices and safety. Our analysis was focused on the ADSCs, rather than the whole stromal vascular fraction (SVF) cell populations, to facilitate characterization that is related to their source of origins. Clinical outcomes improvement suggested that these cells hold great promise in stem cell-based therapies in neurodegenerative, cardiovascular, and auto-immunes diseases.
Collapse
Affiliation(s)
- Loubna Mazini
- Laboratoire Cellules Souches et Ingénierie Tissulaire, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
| | - Luc Rochette
- Equipe d'Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France.
| | - Mohamed Amine
- Laboratoire d'Epidémiologie et de Biostatique, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
- Département de Santé Publique et de Médecine Communautaire, Faculté de Médecine et de Pharmacie, Université Cadi Ayyad, Marrakech 40000, Morocco.
| | - Gabriel Malka
- Laboratoire Cellules Souches et Ingénierie Tissulaire, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
- Laboratoire d'Epidémiologie et de Biostatique, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
| |
Collapse
|
30
|
Mazini L, Rochette L, Amine M, Malka G. Regenerative Capacity of Adipose Derived Stem Cells (ADSCs), Comparison with Mesenchymal Stem Cells (MSCs). Int J Mol Sci 2019. [PMID: 31121953 DOI: 10.3390/ijms20102523.pmid:31121953;pmcid:pmc6566837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
Adipose tissue is now on the top one of stem cell sources regarding its accessibility, abundance, and less painful collection procedure when compared to other sources. The adipose derived stem cells (ADSCs) that it contains can be maintained and expanded in culture for long periods of time without losing their differentiation capacity, leading to large cell quantities being increasingly used in cell therapy purposes. Many reports showed that ADSCs-based cell therapy products demonstrated optimal efficacy and efficiency in some clinical indications for both autologous and allogeneic purposes, hence becoming considered as potential tools for replacing, repairing, and regenerating dead or damaged cells. In this review, we analyzed the therapeutic advancement of ADSCs in comparison to bone marrow (BM) and umbilical cord (UC)-mesenchymal stem cells (MSCs) and designed the specific requirements to their best clinical practices and safety. Our analysis was focused on the ADSCs, rather than the whole stromal vascular fraction (SVF) cell populations, to facilitate characterization that is related to their source of origins. Clinical outcomes improvement suggested that these cells hold great promise in stem cell-based therapies in neurodegenerative, cardiovascular, and auto-immunes diseases.
Collapse
Affiliation(s)
- Loubna Mazini
- Laboratoire Cellules Souches et Ingénierie Tissulaire, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
| | - Luc Rochette
- Equipe d'Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France.
| | - Mohamed Amine
- Laboratoire d'Epidémiologie et de Biostatique, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
- Département de Santé Publique et de Médecine Communautaire, Faculté de Médecine et de Pharmacie, Université Cadi Ayyad, Marrakech 40000, Morocco.
| | - Gabriel Malka
- Laboratoire Cellules Souches et Ingénierie Tissulaire, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
- Laboratoire d'Epidémiologie et de Biostatique, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
| |
Collapse
|
31
|
Oltean-Dan D, Dogaru GB, Apostu D, Mester A, Benea HRC, Paiusan MG, Popa CO, Jianu EM, Bodizs GI, Berce C, Toader AM, Tomoaia G. Enhancement of bone consolidation using high-frequency pulsed electromagnetic fields (HF-PEMFs): An experimental study on rats. Bosn J Basic Med Sci 2019; 19:201-209. [PMID: 30794499 DOI: 10.17305/bjbms.2019.3854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/13/2018] [Indexed: 11/16/2022] Open
Abstract
In vitro studies showed that high-frequency pulsed electromagnetic fields (HF-PEMFs) increase the activity/expression of early and late osteogenic markers and enhance bone mineralization. The main aim of this study was to investigate the in vivo effects of HF-PEMFs on fracture healing using a rat model. A femur fracture was established by surgery in 20 male Wistar rats. Titanium nails were implanted to reduce and stabilize the fracture. After surgery, 20 rats were equally divided into untreated control and treated group (from the first postoperative day HF-PEMFs at 400 pulses/sec [pps] were applied for 10 minutes/day, for two weeks). Quantitative and qualitative assessment of bone formation was made at two and eight weeks following surgery and included morphological and histological analysis, serological analysis by ELISA, micro-computed tomography (micro-CT), and three-point bending test. At two weeks in HF-PEMF group, soft callus was at a more advanced fibrocartilaginous stage and the bone volume/total tissue volume (BV/TV) ratio in the callus area was significantly higher compared to control group (p = 0.047). Serum concentration of alkaline phosphatase (ALP) and osteocalcin (OC) was significantly higher in HF-PEMF group (ALP p = 0.026, OC p = 0.006) as well as the mechanical strength of femurs (p = 0.03). At eight weeks, femurs from HF-PEMF group had a completely formed woven bone with dense trabeculae, active bone marrow, and had a significantly higher BV/TV ratio compared to control (p = 0.01). HF-PEMFs applied from the first postoperative day, 10 minutes/day for two weeks, enhance bone consolidation in rats, especially in the early phase of fracture healing.
Collapse
Affiliation(s)
- Daniel Oltean-Dan
- "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Dual functional approaches for osteogenesis coupled angiogenesis in bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109761. [PMID: 31349418 DOI: 10.1016/j.msec.2019.109761] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/11/2019] [Accepted: 05/15/2019] [Indexed: 12/31/2022]
Abstract
Bone fracture healing is a multistep and overlapping process of inflammation, angiogenesis and osteogenesis. It is initiated by inflammation, causing the release of various cytokines and growth factors. It leads to the recruitment of stem cells and formation of vasculature resulting in the functional bone formation. This combined phenomenon is used by bone tissue engineers from past few years to address the problem of vasculature and osteogenic differentiation during bone regeneration. In this review, we have discussed all major studies reporting the dual functioning approach to promote osteogenesis coupled angiogenesis using various scaffolds. These scaffolds are broadly classified into four types based on the nature of their structural and functional components. The functionality of the scaffold is either due to the structural components or the loaded cargo which conducts or induces the coupled functionality. Dual delivery system for osteoinductive and angioinductive factors ensures the co-delivery of two different types of molecules to induce osteogenesis and angiogenesis. Single delivery scaffold for angioinductive and osteoinductive molecule releases single type of molecules which could induce both angiogenesis and osteogenesis. Osteoconductive scaffold consisted of bone constituents releases angioinductive factors. Osteoconductive and angioconductive scaffold composed of components which provide the native substrate features for osteogenesis and angiogenesis. This review article also discusses the studies highlighting the synergism of physico-chemical stimuli as dual functioning feature to enhance angiogenesis and osteogenesis simultaneously. In addition, this article covers one of the least discussed area of the bone regeneration i.e. 'cartilage formation as a median between angiogenesis and osteogenesis'.
Collapse
|
33
|
González-Gil AB, Lamo-Espinosa JM, Muiños-López E, Ripalda-Cemboráin P, Abizanda G, Valdés-Fernández J, López-Martínez T, Flandes-Iparraguirre M, Andreu I, Elizalde MR, Stuckensen K, Groll J, De-Juan-Pardo EM, Prósper F, Granero-Moltó F. Periosteum-derived mesenchymal progenitor cells in engineered implants promote fracture healing in a critical-size defect rat model. J Tissue Eng Regen Med 2019; 13:742-752. [PMID: 30785671 DOI: 10.1002/term.2821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/01/2019] [Accepted: 02/13/2019] [Indexed: 11/06/2022]
Abstract
An attractive alternative to bone autografts is the use of autologous mesenchymal progenitor cells (MSCs) in combination with biomaterials. We compared the therapeutic potential of different sources of mesenchymal stem cells in combination with biomaterials in a bone nonunion model. A critical-size defect was created in Sprague-Dawley rats. Animals were divided into six groups, depending on the treatment to be applied: bone defect was left empty (CTL); treated with live bone allograft (LBA); hrBMP-2 in collagen scaffold (CSBMP2 ); acellular polycaprolactone scaffold (PCL group); PCL scaffold containing periosteum-derived MSCs (PCLPMSCs ) and PCL containing bone marrow-derived MSCs (PCLBMSCs ). To facilitate cell tracking, both MSCs and bone graft were isolated from green fluorescent protein (GFP)-transgenic rats. CTL group did not show any signs of healing during the radiological follow-up (n = 6). In the LBA group, all the animals showed bone bridging (n = 6) whereas in the CSBMP2 group, four out of six animals demonstrated healing. In PCL and PCLPMSCs groups, a reduced number of animals showed radiological healing, whereas no healing was detected in the PCLBMSCs group. Using microcomputed tomography, the bone volume filling the defect was quantified, showing significant new bone formation in the LBA, CSBMP2 , and PCLPMSCs groups when compared with the CTL group. At 10 weeks, GFP positive cells were detected only in the LBA group and restricted to the outer cortical bone in close contact with the periosteum. Tracking of cellular implants demonstrated significant survival of the PMSCs when compared with BMSCs. In conclusion, PMSCs improve bone regeneration being suitable for mimetic autograft design.
Collapse
Affiliation(s)
- Ana B González-Gil
- Orthopaedic Surgery and Traumatology Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - José M Lamo-Espinosa
- Orthopaedic Surgery and Traumatology Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - Emma Muiños-López
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | | | - Gloria Abizanda
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | - José Valdés-Fernández
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | - Tania López-Martínez
- Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| | | | - Ion Andreu
- TECNUN, Universidad de Navarra, San Sebastian, Spain
| | - María Reyes Elizalde
- TECNUN, Universidad de Navarra, San Sebastian, Spain.,CEIT, San Sebastian, Spain
| | - Kai Stuckensen
- Department of Functional Materials in Medicine and Dentistry, University of Würzburg, Würzburg, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry, University of Würzburg, Würzburg, Germany
| | - Elena M De-Juan-Pardo
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Felipe Prósper
- Orthopaedic Surgery and Traumatology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain.,Hematology and Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain
| | - Froilán Granero-Moltó
- Orthopaedic Surgery and Traumatology Department, Clínica Universidad de Navarra, Pamplona, Spain.,Cell Therapy Area, Centro de Investigación Médica Aplicada, IDISNA, Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
34
|
Houschyar KS, Tapking C, Borrelli MR, Popp D, Duscher D, Maan ZN, Chelliah MP, Li J, Harati K, Wallner C, Rein S, Pförringer D, Reumuth G, Grieb G, Mouraret S, Dadras M, Wagner JM, Cha JY, Siemers F, Lehnhardt M, Behr B. Wnt Pathway in Bone Repair and Regeneration - What Do We Know So Far. Front Cell Dev Biol 2019; 6:170. [PMID: 30666305 PMCID: PMC6330281 DOI: 10.3389/fcell.2018.00170] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/30/2018] [Indexed: 02/05/2023] Open
Abstract
Wnt signaling plays a central regulatory role across a remarkably diverse range of functions during embryonic development, including those involved in the formation of bone and cartilage. Wnt signaling continues to play a critical role in adult osteogenic differentiation of mesenchymal stem cells. Disruptions in this highly-conserved and complex system leads to various pathological conditions, including impaired bone healing, autoimmune diseases and malignant degeneration. For reconstructive surgeons, critically sized skeletal defects represent a major challenge. These are frequently associated with significant morbidity in both the recipient and donor sites. The Wnt pathway is an attractive therapeutic target with the potential to directly modulate stem cells responsible for skeletal tissue regeneration and promote bone growth, suggesting that Wnt factors could be used to promote bone healing after trauma. This review summarizes our current understanding of the essential role of the Wnt pathway in bone regeneration and repair.
Collapse
Affiliation(s)
- Khosrow S Houschyar
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Christian Tapking
- Department of Surgery, Shriners Hospital for Children-Galveston, University of Texas Medical Branch, Galveston, TX, United States.,Department of Hand, Plastic and Reconstructive Surgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Heidelberg, Germany
| | - Mimi R Borrelli
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, United States
| | - Daniel Popp
- Department of Surgery, Shriners Hospital for Children-Galveston, University of Texas Medical Branch, Galveston, TX, United States.,Division of Hand, Plastic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Dominik Duscher
- Department of Plastic Surgery and Hand Surgery, Technical University Munich, Munich, Germany
| | - Zeshaan N Maan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, United States
| | - Malcolm P Chelliah
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, United States
| | - Jingtao Li
- State Key Laboratory of Oral Diseases and Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kamran Harati
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Christoph Wallner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Susanne Rein
- Department of Plastic and Hand Surgery-Burn Center-Clinic St. Georg, Leipzig, Germany
| | - Dominik Pförringer
- Clinic and Policlinic of Trauma Surgery, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Georg Reumuth
- Department of Plastic and Hand Surgery, Burn Unit, Trauma Center Bergmannstrost Halle, Halle, Germany
| | - Gerrit Grieb
- Department of Plastic Surgery and Hand Surgery, Gemeinschaftskrankenhaus Havelhoehe, Teaching Hospital of the Charité Berlin, Berlin, Germany
| | - Sylvain Mouraret
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, CA, United States.,Department of Periodontology, Service of Odontology, Rothschild Hospital, AP-HP, Paris 7 - Denis, Diderot University, U.F.R. of Odontology, Paris, France
| | - Mehran Dadras
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Johannes M Wagner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Jungul Y Cha
- Orthodontic Department, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Frank Siemers
- Department of Plastic and Hand Surgery, Burn Unit, Trauma Center Bergmannstrost Halle, Halle, Germany
| | - Marcus Lehnhardt
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Björn Behr
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
35
|
Besio R, Maruelli S, Battaglia S, Leoni L, Villani S, Layrolle P, Rossi A, Trichet V, Forlino A. Early Fracture Healing is Delayed in the Col1a2 +/G610C Osteogenesis Imperfecta Murine Model. Calcif Tissue Int 2018; 103:653-662. [PMID: 30076439 DOI: 10.1007/s00223-018-0461-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/30/2018] [Indexed: 01/08/2023]
Abstract
Osteogenesis imperfecta (OI) is a rare heritable skeletal dysplasia mainly caused by type I collagen abnormalities and characterized by bone fragility and susceptibility to fracture. Over 85% of the patients carry dominant mutations in the genes encoding for the collagen type I α1 and α2 chains. Failure of bone union and/or presence of hyperplastic callus formation after fracture were described in OI patients. Here we used the Col1a2+/G610C mouse, carrying in heterozygosis the α2(I)-G610C substitution, to investigate the healing process of an OI bone. Tibiae of 2-month-old Col1a2+/G610C and wild-type littermates were fractured and the healing process was followed at 2, 3, and 5 weeks after injury from fibrous cartilaginous tissue formation to its bone replacement by radiography, micro-computed tomography (µCT), histological and biochemical approaches. In presence of similar fracture types, in Col1a2+/G610C mice an impairment in the early phase of bone repair was detected compared to wild-type littermates. Smaller callus area, callus bone surface, and bone volume associated to higher percentage of cartilage and lower percentage of bone were evident in Col1a2+/G610C at 2 weeks post fracture (wpf) and no change by 3 wpf. Furthermore, the biochemical analysis of collagen extracted from callus 2 wpf revealed in mutants an increased amount of type II collagen, typical of cartilage, with respect to type I, characteristic of bone. This is the first report of a delay in OI bone fracture repair at the modeling phase.
Collapse
Affiliation(s)
- Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Silvia Maruelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Severine Battaglia
- INSERM, UMR 1238, PHY-OS, Bone sarcomas and remodeling of calcified tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - Laura Leoni
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Simona Villani
- Department of Public Health and Experimental and Forensic Medicine, Unit of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia, Italy
| | - Pierre Layrolle
- INSERM, UMR 1238, PHY-OS, Bone sarcomas and remodeling of calcified tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - Antonio Rossi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Valerie Trichet
- INSERM, UMR 1238, PHY-OS, Bone sarcomas and remodeling of calcified tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy.
| |
Collapse
|
36
|
Tian L, Sheng Y, Huang L, Chow DHK, Chau WH, Tang N, Ngai T, Wu C, Lu J, Qin L. An innovative Mg/Ti hybrid fixation system developed for fracture fixation and healing enhancement at load-bearing skeletal site. Biomaterials 2018; 180:173-183. [PMID: 30041069 DOI: 10.1016/j.biomaterials.2018.07.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 07/05/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
Magnesium (Mg) is a potential biomaterial suitable for developing biodegradable orthopaedic implants, especially as internal fixators for fracture fixation at non-load bearing skeletal sites. However, Mg alone cannot provide sufficient mechanical support for stable fracture fixation at load bearing sites due to its rapid degradation in the early stage after implantation. In consideration of the strengths and weaknesses of Mg, we developed an innovative magnesium/titanium (Mg/Ti) hybrid fixation system for long bone fracture fixation and investigated the fixation efficacy. The finite element analysis (FEA) results indicated that the Mg/Ti hybrid fixation system provided sufficient mechanical support for fracture fixation at load-bearing skeletal site. As a proof-of-concept, we performed a "Z-shaped" open osteotomy at the mid-shaft of rabbit tibia. For comparison, the animals were divided into two groups: Mg/Ti group (fixated with Mg screws and Ti fixators) and Ti control group (fixated with Ti screws and Ti fixators). The radiographic, four-point bending mechanical test, histological and histomorphometric analysis were postoperatively performed in a temporal manner up to 12 weeks. Both X-ray and micro-CT images of the Mg/Ti group showed a larger callus (14.7% at 3rd week and 24.8% at 6th week, n = 5-7, p < 0.05) in the regions of interest (ROIs) over time, especially at the opposite cortex of the fixation plate. At the 12th week post-operation, the biomechanical test result indicated that the rabbit tibia in the Mg/Ti group healed better and the overall mechanical strength was approximately 3-fold higher (n = 8, p < 0.05) than that at 6th week. Furthermore, the FEA revealed that the Mg/Ti group had a higher mechanical strength (19.5% at week 6 and 31.5% at week 12) at the specified ROI and resulted in an earlier and faster endochondral ossification (68.0% at week 3 and 71.4% at week 6) with a higher expression of osteocalcin (54.0%) and collagen I (34.2%) than the Ti control group (n = 4, p < 0.05). Further evaluation suggested that a higher expression of calcitonin gene-related peptide (CGRP), a known osteogenic neuron peptide, in the fracture callus of the Mg/Ti group might be a major underlying mechanism of enhanced fracture healing attributed to the release of Mg ions during the degradation of Mg screws.
Collapse
Affiliation(s)
- Li Tian
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Yifeng Sheng
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Le Huang
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Dick Ho-Kiu Chow
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Wing Ho Chau
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Ning Tang
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - To Ngai
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Chi Wu
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Jian Lu
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region
| | - Ling Qin
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
37
|
Aziz AUA, Wahab AA, Ramlee MH. Relationship Between Strain and Healing Process for the Use of External Fixator: A Short Review. 2018 2ND INTERNATIONAL CONFERENCE ON BIOSIGNAL ANALYSIS, PROCESSING AND SYSTEMS (ICBAPS) 2018. [DOI: 10.1109/icbaps.2018.8527393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
38
|
Cooke ME, Hussein AI, Lybrand KE, Wulff A, Simmons E, Choi JH, Litrenta J, Ricci WM, Nascone JW, O’Toole RV, Morgan EF, Gerstenfeld LC, Tornetta P. Correlation between RUST assessments of fracture healing to structural and biomechanical properties. J Orthop Res 2018; 36:945-953. [PMID: 28833572 PMCID: PMC5823715 DOI: 10.1002/jor.23710] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/19/2017] [Indexed: 02/04/2023]
Abstract
Radiographic Union Score for Tibia (RUST) and modified RUST (mRUST) are radiographic tools for quantitatively evaluating fracture healing using a cortical scoring system. This tool has high intra-class correlation coefficients (ICCs); however, little evidence has evaluated the scores against the physical properties of bone healing. Closed, stabilized fractures were made in the femora of C3H/HeJ male mice (8-12 week-old) of two dietary groups: A control and a phosphate restricted diet group. Micro-computed tomography (µCT) and torsion testing were carried out at post-operative days (POD) 14, 21, 35, and 42 (n = 10-16) per group time-point. Anteroposterior and lateral radiographic views were constructed from the µCT scans and scored by five raters. The raters also indicated if the fracture were healed. ICCs were 0.71 (mRUST) and 0.63 (RUST). Both RUST scores were positively correlated with callus bone mineral density (BMD) (r = 0.85 and 0.80, p < 0.001) and bone volume fraction (BV/TV) (r = 0.86 and 0.80, p < 0.001). Both RUST scores positively correlated with callus strength (r = 0.35 and 0.26, p < 0.012) and rigidity (r = 0.50 and 0.39, p < 0.001). Radiographically healed calluses had a mRUST ≥13 and a RUST ≥10 and had excellent relationship to structural and biomechanical metrics. Effect of delayed healing due to phosphate dietary restrictions was found at later time points with all mechanical properties (p < 0.011), however no differences found in the RUST scores (p > 0.318). Clinical relevance of this study is both RUST scores showed high correlation to physical properties of healing and generally distinguished healed vs. non-healed fractures. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:945-953, 2018.
Collapse
Affiliation(s)
- Margaret E Cooke
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Amira I Hussein
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Kyle E Lybrand
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Alexander Wulff
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Erin Simmons
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Jeffrey H Choi
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - Jody Litrenta
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| | - William M Ricci
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO
| | - Jason W Nascone
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Robert V O’Toole
- Department of Orthopaedic Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Elise F Morgan
- Department of Mechanical Engineering, Boston University, Boston, MA
| | | | - Paul Tornetta
- Department of Orthopaedic Surgery, Boston University, Boston, MA
| |
Collapse
|
39
|
Effect of a Particulate and a Putty-Like Tricalcium Phosphate-Based Bone-grafting Material on Bone Formation, Volume Stability and Osteogenic Marker Expression after Bilateral Sinus Floor Augmentation in Humans. J Funct Biomater 2017; 8:jfb8030031. [PMID: 28758916 PMCID: PMC5618282 DOI: 10.3390/jfb8030031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 07/25/2017] [Accepted: 07/25/2017] [Indexed: 01/25/2023] Open
Abstract
This study examines the effect of a hyaluronic acid (HyAc) containing tricalcium phosphate putty scaffold material (TCP-P) and of a particulate tricalcium phosphate (TCP-G) graft on bone formation, volume stability and osteogenic marker expression in biopsies sampled 6 months after bilateral sinus floor augmentation (SFA) in 7 patients applying a split-mouth design. 10% autogenous bone chips were added to the grafting material during surgery. The grain size of the TCP granules was 700 to 1400 µm for TCP-G and 125 to 250 µm and 500 to 700 µm (ratio 1:1) for TCP-P. Biopsies were processed for immunohistochemical analysis of resin-embedded sections. Sections were stained for collagen type I (Col I), alkaline phosphatase (ALP), osteocalcin (OC) and bone sialoprotein (BSP). Furthermore, the bone area and biomaterial area fraction were determined histomorphometrically. Cone-beam CT data recorded after SFA and 6 months later were used for calculating the graft volume at these two time points. TCP-P displayed more advantageous surgical handling properties and a significantly greater bone area fraction and smaller biomaterial area fraction. This was accompanied by significantly greater expression of Col I and BSP and in osteoblasts and osteoid and a less pronounced reduction in grafting volume with TCP-P. SFA using both types of materials resulted in formation of sufficient bone volume for facilitating stable dental implant placement with all dental implants having been in function without any complications for 6 years. Since TCP-P displayed superior surgical handling properties and greater bone formation than TCP-G, without the HyAc hydrogel matrix having any adverse effect on bone formation or graft volume stability, TCP-P can be regarded as excellent grafting material for SFA in a clinical setting. The greater bone formation observed with TCP-P may be related to the difference in grain size of the TCP granules and/or the addition of the HyAc.
Collapse
|
40
|
Wang P, Ying J, Luo C, Jin X, Zhang S, Xu T, Zhang L, Mi M, Chen D, Tong P, Jin H. Osthole Promotes Bone Fracture Healing through Activation of BMP Signaling in Chondrocytes. Int J Biol Sci 2017; 13:996-1007. [PMID: 28924381 PMCID: PMC5599905 DOI: 10.7150/ijbs.19986] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/18/2017] [Indexed: 11/16/2022] Open
Abstract
Osthole is a bioactive coumarin derivative and has been reported to be able to enhance bone formation and improve fracture healing. However, the molecular mechanism of Osthole in bone fracture healing has not been fully defined. In this study we determined if Osthole enhances bone fracture healing through activation of BMP2 signaling in mice. We performed unilateral open transverse tibial fracture procedure in 10-week-old C57BL/6 mice which were treated with or without Osthole. Our previous studies demonstrated that chondrocyte BMP signaling is required for bone fracture healing, in this study we also performed tibial fracture procedure in Cre-negative and Col2-Cre;Bmp2flox/flox conditional knockout (KO) mice (Bmp2Col2Cre) to determine if Osthole enhances fracture healing in a BMP2-dependent manner. Fracture callus tissues were collected and analyzed by X-ray, micro-CT (μCT), histology, histomorphometry, immunohistochemistry (IHC), biomechanical testing and quantitative gene expression analysis. In addition, mouse chondrogenic ATDC5 cells were cultured with or without Osthole and the expression levels of chondrogenic marker genes were examined. The results demonstrated that Osthole promotes bone fracture healing in wild-type (WT) or Cre- control mice. In contrast, Osthole failed to promote bone fracture healing in Bmp2Col2Creconditional KO mice. In the mice receiving Osthole treatment, expression of cartilage marker genes was significantly increased. We conclude that Osthole could promote bone strength and enhance fracture healing by activation of BMP2 signaling. Osthole may be used as an alternative approach in the orthopaedic clinic for the treatment of fracture healing.
Collapse
Affiliation(s)
- Pinger Wang
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jun Ying
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,First Clinical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Cheng Luo
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,First Clinical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Xing Jin
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,Department of Orthopaedics and Traumatology, Wangjiang Sub-District Community Health Service Centre, Hangzhou 310016, Zhejiang Province, China
| | - Shanxing Zhang
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Taotao Xu
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,First Clinical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Lei Zhang
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,First Clinical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Meng Mi
- Department of Traumatology, Beijing Jishuitan Hospital, 100035, Beijing, China
| | - Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - Peijian Tong
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China.,Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang Province, China
| |
Collapse
|
41
|
Roth TD, Ladd LM, Kempton LB. Fracture Healing and Imaging Evaluation. CURRENT RADIOLOGY REPORTS 2017. [DOI: 10.1007/s40134-017-0221-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
42
|
Effect of sex-hormone levels, sex, body mass index and other host factors on human craniofacial bone regeneration with bioactive tricalcium phosphate grafts. Biomaterials 2017; 123:48-62. [PMID: 28160669 DOI: 10.1016/j.biomaterials.2017.01.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/23/2016] [Accepted: 01/27/2017] [Indexed: 12/16/2022]
Abstract
Little is known regarding the associations between sex-hormone levels, sex, body mass index (BMI), age, other host factors and biomaterial stimulated bone regeneration in the human craniofacial skeleton. The aim of this study was to elucidate the associations between these factors and bone formation after sinus floor augmentation procedures (SFA) utilizing a bioactive tricalcium phosphate (TCP) bone grafting material. We conducted a prospective study in a human population in which 60 male and 60 female participants underwent SFA and dental implant placement using a staged approach. BMI as well as levels of serum estradiol (E2), total testosterone (TT), and the free androgen index (FAI) were measured by radioimmunoassay and electrochemoluminescent-immunoassay. At implant placement, 6 months after SFA, bone biopsy specimens were harvested for hard tissue histology, the amount of bone formation was evaluated by histomorphometry and immunohistochemical analysis of osteogenic marker expression. The Wilcoxon rank-sum U test, Spearman correlations and linear regression analysis were used to explore the association between bone formation and BMI, hormonal and other host factors. BMI and log E2 were significantly positively associated with bone formation in male individuals (p < 0.05). Histomorphometry revealed trends toward greater bone formation and osteogenic marker expression with non-smokers compared to smokers. In male patients, higher E2 levels and higher BMI enhanced TCP stimulated craniofacial i.e. intramembranous bone repair.
Collapse
|
43
|
Abstract
Mechanoresponses in mesenchymal stem cells (MSCs) guide both differentiation and function. In this review, we focus on advances in0 our understanding of how the cytoplasmic cytoskeleton, nuclear envelope and nucleoskeleton, which are connected via LINC (Linker of Nucleoskeleton and Cytoskeleton) complexes, are emerging as an integrated dynamic signaling platform to regulate MSC mechanobiology. This dynamic interconnectivity affects mechanical signaling and transfer of signals into the nucleus. In this way, nuclear and LINC-mediated cytoskeletal connectivity play a critical role in maintaining mechanical signaling that affects MSC fate by serving as both mechanosensory and mechanoresponsive structures. We review disease and age related compromises of LINC complexes and nucleoskeleton that contribute to the etiology of musculoskeletal diseases. Finally we invite the idea that acquired dysfunctions of LINC might be a contributing factor to conditions such as aging, microgravity and osteoporosis and discuss potential mechanical strategies to modulate LINC connectivity to combat these conditions.
Collapse
|
44
|
Féron JM, Mauprivez R. Fracture repair: general aspects and influence of osteoporosis and anti-osteoporosis treatment. Injury 2016; 47 Suppl 1:S10-4. [PMID: 26768282 DOI: 10.1016/s0020-1383(16)30003-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Bone differs from other tissues in its capacity to self-repair after a fracture. The low bone mass and structural deterioration of bone associated with osteoporosis increases the risk of fragility fracture compared with healthy individuals. The intention of this article is to review the complex process of fracture repair and essential requirements for a successful fracture healing response summarized as the "diamond concept" in terms of aging and osteoporosis. The current preclinical and clinical evidence for a beneficial or harmful influence of anti-osteoporosis medications such as bisphosphonates, parathyroid hormone (PTH), strontium ranelate and antibodies of Wnt-inhibiting signaling proteins on bone healing is presented and discussed. Literature suggests that there are no detrimental consequences of such therapeutics on fracture repair processes. Following a fragility fracture, it seems that early start of preventive anti-osteoporotic treatment right after surgery does not delay the union of the fracture, except perhaps in the case of very rigidly fixed fracture requiring direct bone healing. There is some promising experimental and clinical evidence for possible enhancement of the bone repair process via administration of systemic agents. Further well designed studies in humans are necessary to accumulate more evidence on the positive effects and to translate this knowledge into valid therapeutic applications.
Collapse
Affiliation(s)
- Jean-Marc Féron
- Professor of Orthopaedic Surgery, Chair of the Orthopaedic and Trauma Department, Saint Antoine Hospital. UPMC-Sorbonne Universities, Paris, France.
| | - Raphaël Mauprivez
- Senior lecturer, Orthopaedic and Trauma Department, Saint Antoine Hospital. UPMC-Sorbonne Universities, Paris, France
| |
Collapse
|