151
|
Kos O, Alexander C, Brandenburg K, Chen Z, Heini A, Heumann D, Khatri I, Mach JP, Rietschel ET, Terskikh A, Ulmer AJ, Waelli T, Yu K, Zähringer U, Gorczynski RM. Regulation of fetal hemoglobin expression during hematopoietic stem cell development and its importance in bone metabolism and osteoporosis. Int Immunopharmacol 2018; 57:112-120. [PMID: 29477972 DOI: 10.1016/j.intimp.2018.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/07/2018] [Accepted: 02/13/2018] [Indexed: 11/19/2022]
Abstract
We have shown that an altered tissue redox environment in mice lacking either murine beta Hemoglobin major (HgbβmaKO) or minor (HgbβmiKO) regulates inflammation. The REDOX environment in marrow stem cell niches also control differentiation pathways. We investigated osteoclastogenesis (OC)/osteoblastogenesis (OB), in bone cultures derived from untreated or FSLE-treated WT, HgbβmaKO or HgbβmiKO mice. Marrow mesenchymal cells from 10d pre-cultures were incubated on an osteogenic matrix for 21d prior to analysis of inflammatory cytokine release into culture supernatants, and relative OC:OB using (TRAP:BSP, RANKL:OPG) mRNA expression ratios and TRAP or Von Kossa staining. Cells from WT and HgbβmaKO mice show decreased IL-1β,TNFα and IL-6 production and enhanced osteoblastogenesis with altered mRNA expression ratios and increased bone nodules (Von Kossa staining) in vitro after in vivo stimulation of mRNA expression of fetal Hgb genes (Hgbε and Hgbβmi) by a fetal liver extract (FSLE). Marrow from HgbβmiKO showed enhanced cytokine release and preferential enhanced osteoclastogenesis relative to similar cells from WT or HgbβmaKO mice, with no increased osteoblastogenesis after mouse treatment with FSLE. Pre-treatment of WT or HgbβmaKO, but not HgbβmiKO mice, with other molecules (rapamycin; hydroxyurea) which increase expression of fetal Hgb genes also augmented osteoblastogenesis and decreased cytokine production in cells differentiating in vitro. Infusion of rabbit anti- Hgbε or anti- Hgbβmi, but not anti-Hgbα or anti- Hgbβma into WT mice from day 13 gestation for 3 weeks led to attenuated osteoblastogenesis in cultured cells. We conclude that increased fetal hemoglobin expression, or use of agents which improve fetal hemoglobin expression, increases osteoblast bone differentiation in association with decreased inflammatory cytokine release.
Collapse
Affiliation(s)
- O Kos
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - C Alexander
- Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - K Brandenburg
- Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - Z Chen
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - A Heini
- Clinique La Prairie, Clarens-Montreux, Switzerland
| | - D Heumann
- Clinique La Prairie, Clarens-Montreux, Switzerland
| | - I Khatri
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - J P Mach
- Department of Biochemistry, University of Lausanne, Switzerland
| | | | - A Terskikh
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - A J Ulmer
- Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - T Waelli
- Clinique La Prairie, Clarens-Montreux, Switzerland
| | - K Yu
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
| | - U Zähringer
- Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - R M Gorczynski
- Institute of Medical Sciences, University of Toronto, Toronto, Canada.
| |
Collapse
|
152
|
Shiu HT, Leung PC, Ko CH. The roles of cellular and molecular components of a hematoma at early stage of bone healing. J Tissue Eng Regen Med 2018; 12:e1911-e1925. [PMID: 29207216 DOI: 10.1002/term.2622] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 10/23/2017] [Accepted: 11/22/2017] [Indexed: 12/14/2022]
Abstract
Bone healing is a complex repair process that commences with the formation of a blood clot at the injured bone, termed hematoma. It has evidenced that a lack of a stable hematoma causes delayed bone healing or non-union. The hematoma at the injured bone constitutes the early healing microenvironment. It appears to dictate healing pathways that ends in a regenerative bone. However, the hematoma is often clinically removed from the damaged site. Conversely, blood-derived products have been used in bone tissue engineering for treating critical sized defects, including fibrin gels and platelet-rich plasma. A second generation of platelet concentrate that is based on leukocyte and fibrin content has also been developed and introduced in market. Conflicting effect of these products in bone repair are reported. We propose that the bone healing response becomes dysregulated if the blood response and subsequent formation and properties of a hematoma are altered. This review focuses on the central structural, cellular, and molecular components of a fracture hematoma, with a major emphasis on their roles in regulating bone healing mechanism, and their interactions with mesenchymal stem cells. New angles towards a better understanding of these factors and relevant mechanisms involved at the beginning of bone healing may help to clarify limited or adverse effects of blood-derived products on bone repair. We emphasize that the recreation of an early hematoma niche with critical compositions might emerge as a viable therapeutic strategy for enhanced skeletal tissue engineering.
Collapse
Affiliation(s)
- Hoi Ting Shiu
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,State Key Laboratory of Phytochemistry & Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Ping Chung Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,State Key Laboratory of Phytochemistry & Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Chun Hay Ko
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.,State Key Laboratory of Phytochemistry & Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| |
Collapse
|
153
|
Wei F, Xiao Y. Modulation of the Osteoimmune Environment in the Development of Biomaterials for Osteogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:69-86. [DOI: 10.1007/978-981-13-0947-2_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
154
|
Chen Z, Bachhuka A, Wei F, Wang X, Liu G, Vasilev K, Xiao Y. Nanotopography-based strategy for the precise manipulation of osteoimmunomodulation in bone regeneration. NANOSCALE 2017; 9:18129-18152. [PMID: 29143002 DOI: 10.1039/c7nr05913b] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Immune cells play vital roles in regulating bone dynamics. Successful bone regeneration requires a favourable osteo-immune environment. The high plasticity and diversity of immune cells make it possible to manipulate the osteo-immune response of immune cells, thus modulating the osteoimmune environment and regulating bone regeneration. With the advancement in nanotechnology, nanotopographies with different controlled surface properties can be fabricated. On tuning the surface properties, the osteo-immune response can be precisely modulated. This highly tunable characteristic and immunomodulatory effects make nanotopography a promising strategy to precisely manipulate osteoimmunomdulation for bone tissue engineering applications. This review first summarises the effects of the immune response during bone healing to show the importance of regulating the immune response for the bone response. The plasticity of immune cells is then reviewed to provide rationales for manipulation of the osteoimmune response. Subsequently, we highlight the current types of nanotopographies applied in bone biomaterials and their fabrication techniques, and explain how these nanotopographies modulate the immune response and the possible underlying mechanisms. The effects of immune cells on nanotopography-mediated osteogenesis are emphasized, and we propose the concept of "nano-osteoimmunomodulation" to provide a valuable strategy for the development of nanotopographies with osteoimmunomodulatory properties that can precisely regulate bone dynamics.
Collapse
Affiliation(s)
- Zetao Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, Guangdong, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
155
|
Walters G, Pountos I, Giannoudis PV. The cytokines and micro-environment of fracture haematoma: Current evidence. J Tissue Eng Regen Med 2017; 12:e1662-e1677. [PMID: 29047220 DOI: 10.1002/term.2593] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 08/31/2017] [Accepted: 10/09/2017] [Indexed: 12/18/2022]
Abstract
Fracture haematoma formation is the first and foremost important stage of fracture healing. It orchestrates the inflammatory and cellular processes leading to the formation of callus and the restoration of the continuity of the bone. Evidence suggests that blocking this initial stage could lead to an impairment of the overall bone healing process. This review aims to analyse the existing evidence of molecular contributions to bone healing within fracture haematoma and to determine the potential to modify the molecular response to fracture in the haematoma with the aim of improving union times. A comprehensive search of literature documenting fracture haematoma cytokine content was performed. Suitable papers according to prespecified criteria were identified and analysed according to Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. A total of 89 manuscripts formed the basis of this analysis. Low oxygen tension, high acidity, and high calcium characterised initially the fracture haematoma micro-environment. In addition, a number of cytokines have been measured with concentrations significantly higher than those found in peripheral circulation. Growth factors have also been isolated, with an observed increase in bone morphogenetic proteins, platelet-derived growth factor, and transforming growth factor. Although molecular modification of fracture haematoma has been attempted, more research is required to determine a suitable biological response modifier leading to therapeutic effects. The cytokine content of fracture haematoma gives insight into processes occurring in the initial stages of fracture healing. Manipulation of signalling molecules represents a promising pathway to target future therapies aiming to upregulate the osteogenesis.
Collapse
Affiliation(s)
- Gavin Walters
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds, UK
| | - Ippokratis Pountos
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds, UK
| | - Peter V Giannoudis
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Unit, Chapel Allerton Hospital, Leeds, UK
| |
Collapse
|
156
|
Prystaz K, Kaiser K, Kovtun A, Haffner-Luntzer M, Fischer V, Rapp AE, Liedert A, Strauss G, Waetzig GH, Rose-John S, Ignatius A. Distinct Effects of IL-6 Classic and Trans-Signaling in Bone Fracture Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:474-490. [PMID: 29146294 DOI: 10.1016/j.ajpath.2017.10.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/28/2017] [Accepted: 10/19/2017] [Indexed: 12/21/2022]
Abstract
Bone healing is a complex process with closely linked phases of inflammation, regeneration, and remodeling. IL-6 may crucially regulate this process; however, the underlying mechanisms are unclear. IL-6 signals are transmitted via the transmembrane glycoprotein 130 by two distinct mechanisms: classic signaling using the membrane-anchored IL-6 receptor and trans-signaling using its soluble form. Herein, we investigated the hypothesis that IL-6 classic and trans-signaling have different functions during bone healing. To investigate fracture healing, 12-week-old C57BL/6J mice underwent a femur osteotomy. To study the function of IL-6 during the inflammatory phase, either an anti-IL-6 antibody, which inhibits IL-6 classic and trans-signaling, or soluble glycoprotein 130 fusion protein, which selectively blocks trans-signaling, was injected after 30 minutes and 48 hours. To analyze IL-6 effects in the repair phase, compounds were injected from day 7 onwards. Global IL-6 inhibition in the early phase after fracture reduced systemic inflammation, the recruitment of immune cells, and bone regeneration, resulting in delayed fracture healing. Global IL-6 inhibition during the repair phase disturbed bone formation and remodeling. In contrast, inhibition of IL-6 trans-signaling exerted minor effects on the immune response and did not influence bone repair, suggesting that the classic pathway accounts for most of the effects observed after global IL-6 inhibition. Our results reveal that IL-6 classic signaling, but not IL-6 trans-signaling, is essential for bone repair.
Collapse
Affiliation(s)
- Katja Prystaz
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, Germany
| | - Kathrin Kaiser
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, Germany
| | - Anna Kovtun
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, Germany
| | | | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, Germany
| | - Anna E Rapp
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, Germany
| | - Astrid Liedert
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, Germany
| | - Gudrun Strauss
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | | | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, Germany.
| |
Collapse
|
157
|
Jung HD, Kim SY, Jung HS, Park HS, Jung YS. Immunohistochemical Analysis on Cortex-to-Cortex Healing After Mandibular Vertical Ramus Osteotomy: A Preliminary Study. J Oral Maxillofac Surg 2017; 76:437.e1-437.e8. [PMID: 29112826 DOI: 10.1016/j.joms.2017.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/05/2017] [Accepted: 10/05/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE The present study analyzed the expression of specific cytokines in the transforming growth factor (TGF)-β superfamily postoperatively after mandibular vertical ramus osteotomy (VRO). MATERIALS AND METHODS Four beagle dogs were enrolled and euthanized at 1, 2, 4, and 8 weeks postoperatively for immunohistochemical analysis using 6 specific antibodies (bone morphogenetic protein [BMP]-2/4, BMP-7, TGF-β2, TGF-β3, matrix metalloproteinase-3, and vascular endothelial growth factor [VEGF]). The results from the surgical site and control (adjacent area) were compared. RESULTS Generalized upregulation of BMP-2/4 was observed in all healing periods, and the strongest expression of BMP-7 was observed at 1 week postoperatively. The strongest expression of TGF-β2 was observed at 8 weeks with increasing pattern. The strong expression of TGF-β3 was observed at 1 and 4 weeks, with the strongest expression of VEGF at 1 week, with a decreasing pattern. No notable uptake was detected with the 6 specific antibodies in the adjacent bone (control). CONCLUSIONS The absence of internal fixation after VRO led to dynamic healing with a specific expression pattern of BMP-7 and TGF-β2. The anatomic factors, including sufficient preexisting vascularity, led to the earlier expression pattern of VEGF.
Collapse
Affiliation(s)
- Hwi-Dong Jung
- Assistant Professor, Department of Oral and Maxillofacial Surgery, Oral Science Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Sang Yoon Kim
- Private Practice, McLean, VA; Former Resident, Harvard Oral and Maxillofacial Surgery, Boston, MA
| | - Han-Sung Jung
- Professor, Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Hyung-Sik Park
- Professor, Department of Oral and Maxillofacial Surgery, Oral Science Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Young-Soo Jung
- Emeritus Professor, Department of Oral and Maxillofacial Surgery, Oral Science Research Institute, Yonsei University College of Dentistry, Seoul, Korea.
| |
Collapse
|
158
|
Bridging hard callus at 48 days in an open femoral shaft fracture with segmental defect treated with a first-stage Masquelet technique: I wasn't expecting that. Strategies Trauma Limb Reconstr 2017; 13:57-60. [PMID: 29116576 PMCID: PMC5862707 DOI: 10.1007/s11751-017-0300-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 10/29/2017] [Indexed: 11/29/2022] Open
Abstract
The Masquelet technique is a strategy for management of segmental bone defects. It is a two-stage procedure that involves inducing a synovial-like membrane that can be used for a bone graft. Segmental bone defects can occur following trauma and can accompany traumatic brain injury. There is a well-documented, albeit debated, association between traumatic brain injury and increased rate of new bone formation. Here, we present a case of unexpected callus formation in a segmental femoral fracture. The patient had a traumatic brain injury and was treated with the first stage of the Masquelet technique. Owing to the amount of large callus, a second stage of the Masquelet was not required. The patient recovered well from the injury and at 16-week follow-up was able to partially weight bear. A case similar to this has not previously been reported within the literature.
Collapse
|
159
|
Graney PL, Roohani-Esfahani SI, Zreiqat H, Spiller KL. In vitro response of macrophages to ceramic scaffolds used for bone regeneration. J R Soc Interface 2017; 13:rsif.2016.0346. [PMID: 27466438 DOI: 10.1098/rsif.2016.0346] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/04/2016] [Indexed: 01/09/2023] Open
Abstract
Macrophages, the primary cells of the inflammatory response, are major regulators of healing, and mediate both bone fracture healing and the inflammatory response to implanted biomaterials. However, their phenotypic contributions to biomaterial-mediated bone repair are incompletely understood. Therefore, we used gene expression and protein secretion analysis to investigate the interactions in vitro between primary human monocyte-derived macrophages and ceramic scaffolds that have been shown to have varying degrees of success in promoting bone regeneration in vivo Specifically, baghdadite (Ca3ZrSi2O9) and strontium-hardystonite-gahnite (Sr-Ca2ZnSi2O7-ZnAl2O4) scaffolds were chosen as two materials that enhanced bone regeneration in vivo in large defects under load compared with clinically used tricalcium phosphate-hydroxyapatite (TCP-HA). Principal component analysis revealed that the scaffolds differentially regulated macrophage phenotype. Temporal changes in gene expression included shifts in markers of pro-inflammatory M1, anti-inflammatory M2a and pro-remodelling M2c macrophage phenotypes. Of note, TCP-HA scaffolds promoted upregulation of many M1-related genes and downregulation of many M2a- and M2c-related genes. Effects of the scaffolds on macrophages were attributed primarily to direct cell-scaffold interactions because of only minor changes observed in transwell culture. Ultimately, elucidating macrophage-biomaterial interactions will facilitate the design of immunomodulatory biomaterials for bone repair.
Collapse
Affiliation(s)
- Pamela L Graney
- Biomaterials and Regenerative Medicine Laboratory, School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Seyed-Iman Roohani-Esfahani
- Biomaterials and Tissue Engineering Research Unit, School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, New South Wales 2026, Australia
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, New South Wales 2026, Australia
| | - Kara L Spiller
- Biomaterials and Regenerative Medicine Laboratory, School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| |
Collapse
|
160
|
Kapasa ER, Giannoudis PV, Jia X, Hatton PV, Yang XB. The Effect of RANKL/OPG Balance on Reducing Implant Complications. J Funct Biomater 2017; 8:E42. [PMID: 28937598 PMCID: PMC5748549 DOI: 10.3390/jfb8040042] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/14/2017] [Accepted: 09/19/2017] [Indexed: 12/27/2022] Open
Abstract
Despite the phenomenal success of implants particularly in the realms of dentistry and orthopaedics, there are still challenges to overcome. The failure of implants resulting from infection, prosthetic loosening, and non-union continue to be the most notorious examples. The cascade of fracture healing and bone repair, especially with the presence of an implant, is complex because it involves a multifaceted immune response alongside the intricate process of bone formation and remodelling. Bone loss is a serious clinical problem that is frequently accompanied by chronic inflammation, illustrating that there is a convoluted relationship between inflammation and bone erosion. The effects of pro-inflammatory factors play a significant role in initiating and maintaining osteoclastogenesis that results in bone resorption by osteoclasts. This is because there is a disruption of the relative ratio between Receptor Activator of Nuclear Factor κB-Ligand (RANKL) and osteoprotegerin (OPG), which is central to modulating bone repair and remodelling. This review aims to provide a background to the bone remodelling process, the bone repair cascade post-implantation, and the associated complications. Furthermore, current clinical solutions that can influence bone formation via either internal or extrinsic mechanisms will be described. These efficacious treatments for osteolysis via targeting the RANKL/OPG ratio may be crucial to reducing the incidence of related implant failures in the future.
Collapse
Affiliation(s)
- Elizabeth R Kapasa
- Doctoral Training Centre-Regenerative Medicine, Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, Leeds LS2 9JT, UK.
- Biomaterials and Tissue Engineering Group, School of Dentistry, University of Leeds, Leeds LS2 9JT, UK.
| | - Peter V Giannoudis
- Department of Trauma and Orthopaedic Surgery, School of Medicine, University of Leeds, Leeds LS2 9JT, UK.
| | - Xiaodong Jia
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK.
| | - Paul V Hatton
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK.
| | - Xuebin B Yang
- Doctoral Training Centre-Regenerative Medicine, Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, Leeds LS2 9JT, UK.
- Biomaterials and Tissue Engineering Group, School of Dentistry, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
161
|
Ivanusic JJ. Molecular Mechanisms That Contribute to Bone Marrow Pain. Front Neurol 2017; 8:458. [PMID: 28955292 PMCID: PMC5601959 DOI: 10.3389/fneur.2017.00458] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/18/2017] [Indexed: 12/11/2022] Open
Abstract
Pain associated a bony pathology puts a significant burden on individuals, society, and the health-care systems worldwide. Pathology that involves the bone marrow activates sensory nerve terminal endings of peripheral bone marrow nociceptors, and is the likely trigger for pain. This review presents our current understanding of how bone marrow nociceptors are influenced by noxious stimuli presented in pathology associated with bone marrow. A number of ion channels and receptors are emerging as important modulators of the activity of peripheral bone marrow nociceptors. Nerve growth factor (NGF) sequestration has been trialed for the management of inflammatory bone pain (osteoarthritis), and there is significant evidence for interaction of NGF with bone marrow nociceptors. Activation of transient receptor potential cation channel subfamily V member 1 sensitizes bone marrow nociceptors and could contribute to increased sensitivity of patients to noxious stimuli in various bony pathologies. Acid-sensing ion channels sense changes to tissue pH in the bone marrow microenvironment and could be targeted to treat pathology that involves acidosis of the bone marrow. Piezo2 is a mechanically gated ion channel that has recently been reported to be expressed by most myelinated bone marrow nociceptors and might be a target for treatments directed against mechanically induced bone pain. These ion channels and receptors could be useful targets for the development of peripherally acting drugs to treat pain of bony origin.
Collapse
Affiliation(s)
- Jason J Ivanusic
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
162
|
Novince CM, Whittow CR, Aartun JD, Hathaway JD, Poulides N, Chavez MB, Steinkamp HM, Kirkwood KA, Huang E, Westwater C, Kirkwood KL. Commensal Gut Microbiota Immunomodulatory Actions in Bone Marrow and Liver have Catabolic Effects on Skeletal Homeostasis in Health. Sci Rep 2017; 7:5747. [PMID: 28720797 PMCID: PMC5515851 DOI: 10.1038/s41598-017-06126-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 06/08/2017] [Indexed: 12/11/2022] Open
Abstract
Despite knowledge the gut microbiota regulates bone mass, mechanisms governing the normal gut microbiota’s osteoimmunomodulatory effects on skeletal remodeling and homeostasis are unclear in the healthy adult skeleton. Young adult specific-pathogen-free and germ-free mice were used to delineate the commensal microbiota’s immunoregulatory effects on osteoblastogenesis, osteoclastogenesis, marrow T-cell hematopoiesis, and extra-skeletal endocrine organ function. We report the commensal microbiota has anti-anabolic effects suppressing osteoblastogenesis and pro-catabolic effects enhancing osteoclastogenesis, which drive bone loss in health. Suppression of Sp7(Osterix) and Igf1 in bone, and serum IGF1, in specific-pathogen-free mice suggest the commensal microbiota’s anti-osteoblastic actions are mediated via local disruption of IGF1-signaling. Differences in the RANKL/OPG Axis in vivo, and RANKL-induced maturation of osteoclast-precursors in vitro, indicate the commensal microbiota induces sustained changes in RANKL-mediated osteoclastogenesis. Candidate mechanisms mediating commensal microbiota’s pro-osteoclastic actions include altered marrow effector CD4+T-cells and a novel Gut-Liver-Bone Axis. The previously unidentified Gut-Liver-Bone Axis intriguingly implies the normal gut microbiota’s osteoimmunomodulatory actions are partly mediated via immunostimulatory effects in the liver. The molecular underpinnings defining commensal gut microbiota immunomodulatory actions on physiologic bone remodeling are highly relevant in advancing the understanding of normal osteoimmunological processes, having implications for the prevention of skeletal deterioration in health and disease.
Collapse
Affiliation(s)
- Chad M Novince
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA.
| | - Carolyn R Whittow
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Johannes D Aartun
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Jessica D Hathaway
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Nicole Poulides
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Michael B Chavez
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Heidi M Steinkamp
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Kaeleigh A Kirkwood
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Emily Huang
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Caroline Westwater
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA.,Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| | - Keith L Kirkwood
- Department of Oral Health Sciences and Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, 29425, USA.,Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, 29425, USA
| |
Collapse
|
163
|
Biology of Bone Formation, Fracture Healing, and Distraction Osteogenesis. J Craniofac Surg 2017; 28:1380-1389. [DOI: 10.1097/scs.0000000000003625] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
164
|
Bara JJ, Dresing I, Zeiter S, Anton M, Daculsi G, Eglin D, Nehrbass D, Stadelmann VA, Betts DC, Müller R, Alini M, Stoddart MJ. A doxycycline inducible, adenoviral bone morphogenetic protein-2 gene delivery system to bone. J Tissue Eng Regen Med 2017; 12:e106-e118. [PMID: 27957814 DOI: 10.1002/term.2393] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/06/2016] [Accepted: 12/06/2016] [Indexed: 12/19/2022]
Abstract
We report the novel use of a tuneable, non-integrating viral gene delivery system to bone that can be combined with clinically approved biomaterials in an 'off-the-shelf' manner. Specifically, a doxycycline inducible Tet-on adenoviral vector (AdTetBMP-2) in combination with mesenchymal stromal cells (MSCs), fibrin and a biphasic calcium phosphate ceramic (MBCP®) was used to repair large bone defects in nude rats. Bone morphogenetic protein-2 (BMP-2) transgene expression could be effectively tuned by modification of the doxycycline concentration. The effect of adenoviral BMP-2 gene delivery upon bone healing was investigated in vivo in 4 mm critically sized, internally fixated, femoral defects. MSCs were transduced either by direct application of AdTetBMP-2 or by pre-coating MBCP granules with the virus. Radiological assessment scores post-mortem were significantly improved upon delivery of AdTetBMP-2. In AdTetBMP-2 groups, histological analysis revealed significantly more newly formed bone at the defect site compared with controls. Newly formed bone was vascularized and fully integrated with nascent tissue and implanted biomaterial. Improvement in healing outcome was achieved using both methods of vector delivery (direct application vs. pre-coating MCBP). Adenoviral delivery of BMP-2 enhanced bone regeneration achieved by the transplantation of MSCs, fibrin and MBCP in vivo. Importantly, our in vitro and in vivo data suggest that this can be achieved with relatively low (ng/ml) levels of the growth factor. Our model and novel gene delivery system may provide a powerful standardized tool for the optimization of growth factor delivery and release for the healing of large bone defects. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | - Iska Dresing
- AO Research Institute Davos, Davos Platz, Switzerland
| | | | - Martina Anton
- Klinikum Rechts der Isar der Technischen Universität München, Institute of Experimental Oncology and Therapy Research, Munich, Germany
| | - Guy Daculsi
- INSERM U791 Laboratory for Osteoarticular and Dental Tissue Engineering, Dental Faculty, Nantes University, Nantes, France
| | - David Eglin
- AO Research Institute Davos, Davos Platz, Switzerland
| | - Dirk Nehrbass
- AO Research Institute Davos, Davos Platz, Switzerland
| | | | - Duncan C Betts
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Davos Platz, Switzerland
| | | |
Collapse
|
165
|
Gene expression profiling of bone marrow mesenchymal stem cells from Osteogenesis Imperfecta patients during osteoblast differentiation. Eur J Med Genet 2017; 60:326-334. [DOI: 10.1016/j.ejmg.2017.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 03/22/2017] [Accepted: 04/05/2017] [Indexed: 12/26/2022]
|
166
|
Lim JC, Ko KI, Mattos M, Fang M, Zhang C, Feinberg D, Sindi H, Li S, Alblowi J, Kayal RA, Einhorn TA, Gerstenfeld LC, Graves DT. TNFα contributes to diabetes impaired angiogenesis in fracture healing. Bone 2017; 99:26-38. [PMID: 28285015 PMCID: PMC5563392 DOI: 10.1016/j.bone.2017.02.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 01/05/2017] [Accepted: 02/26/2017] [Indexed: 02/07/2023]
Abstract
Diabetes increases the likelihood of fracture, interferes with fracture healing and impairs angiogenesis. The latter may be significant due to the critical nature of angiogenesis in fracture healing. Although it is known that diabetes interferes with angiogenesis the mechanisms remain poorly defined. We examined fracture healing in normoglycemic and streptozotocin-induced diabetic mice and quantified the degree of angiogenesis with antibodies to three different vascular markers, CD34, CD31 and Factor VIII. The role of diabetes-enhanced inflammation was investigated by treatment of the TNFα-specific inhibitor, pegsunercept starting 10days after induction of fractures. Diabetes decreased both angiogenesis and VEGFA expression by chondrocytes. The reduced angiogenesis and VEGFA expression in diabetic fractures was rescued by specific inhibition of TNF in vivo. In addition, the TNF inhibitor rescued the negative effect of diabetes on endothelial cell proliferation and endothelial cell apoptosis. The effect of TNFα in vitro was enhanced by high glucose and an advanced glycation endproduct to impair microvascular endothelial cell proliferation and tube formation and to stimulate apoptosis. The effect of TNF, high glucose and an AGE was mediated by the transcription factor FOXO1, which increased expression of p21 and caspase-3. These studies indicate that inflammation plays a major role in diabetes-impaired angiogenesis in endochondral bone formation through its effect on microvascular endothelial cells and FOXO1.
Collapse
Affiliation(s)
- Jason C Lim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kang I Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcelo Mattos
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Miao Fang
- Department of Endocrinology, Shanxi Province People's Hospital, Shanxi Province, China
| | - Citong Zhang
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Implantology, School of Stomatology, Jilin University, Changchun 130021, China
| | - Daniel Feinberg
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hisham Sindi
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuai Li
- Department of Implant Dentistry, Peking University, School and Hospital of Stomatology, Beijing, China
| | - Jazia Alblowi
- Department of Oral Basic and Clinical Sciences, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rayyan A Kayal
- Department of Oral Basic and Clinical Sciences, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Thomas A Einhorn
- Department of Orthopedic Surgery, School of Medicine, Boston University, Boston, MA 02118, USA
| | - Louis C Gerstenfeld
- Department of Orthopedic Surgery, School of Medicine, Boston University, Boston, MA 02118, USA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
167
|
Su YW, Zhou XF, Foster BK, Grills BL, Xu J, Xian CJ. Roles of neurotrophins in skeletal tissue formation and healing. J Cell Physiol 2017; 233:2133-2145. [PMID: 28370021 DOI: 10.1002/jcp.25936] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 12/21/2022]
Abstract
Neurotrophins and their receptors are key molecules that are known to be critical in regulating nervous system development and maintenance and have been recognized to be also involved in regulating tissue formation and healing in skeletal tissues. Studies have shown that neurotrophins and their receptors are widely expressed in skeletal tissues, implicated in chondrogenesis, osteoblastogenesis, and osteoclastogenesis, and are also involved in regulating tissue formation and healing events in skeletal tissue. Increased mRNA expression for neurotrophins NGF, BDNF, NT-3, and NT-4, and their Trk receptors has been observed in injured bone tissues, and NT-3 and its receptor, TrkC, have been identified to have the highest induction at the injury site in a drill-hole injury repair model in both bone and the growth plate. In addition, NT-3 has also recently been shown to be both an osteogenic and angiogenic factor, and this neurotrophin can also enhance expression of the key osteogenic factor, BMP-2, as well as the major angiogenic factor, VEGF, to promote bone formation, vascularization, and healing of the injury site. Further studies, however, are needed to investigate if different neurotrophins have differential roles in skeletal repair, and if NT-3 can be a potential target of intervention for promoting bone fracture healing.
Collapse
Affiliation(s)
- Yu-Wen Su
- Sansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Xin-Fu Zhou
- Sansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Bruce K Foster
- Department of Orthopaedic Surgery, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Brian L Grills
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Cory J Xian
- Sansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
168
|
Abstract
Bone healing involves complex biological pathways and interactions among various cell types and microenvironments. Among them, the monocyte–macrophage–osteoclast lineage and the mesenchymal stem cell–osteoblast lineage are critical, in addition to an initial inflammatory microenvironment. These cellular interactions induce the necessary inflammatory milieu and provide the cells for bone regeneration and immune modulation. Increasing age is accompanied with a rise in the basal state of inflammation, potentially impairing osteogenesis. The translational potential of this article: Translational research has shown multiple interactions between inflammation, ageing, and bone regeneration. This review presents recent, relevant considerations regarding the effects of inflammation and ageing on bone healing.
Collapse
Affiliation(s)
- Emmanuel Gibon
- Corresponding author. Department of Orthopaedic Surgery, Stanford University, 300 Pasteur Drive, Edwards Building R116, Stanford, CA 94305, USA.Department of Orthopaedic SurgeryStanford University300 Pasteur DriveEdwards Building R116StanfordCA94305USA
| | | | | | | |
Collapse
|
169
|
Delayed Endothelial Progenitor Cell Therapy Promotes Bone Defect Repair in a Clinically Relevant Rat Model. Stem Cells Int 2017; 2017:7923826. [PMID: 28523072 PMCID: PMC5390645 DOI: 10.1155/2017/7923826] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 02/14/2017] [Indexed: 11/18/2022] Open
Abstract
The repair of segmental bone defects remains a significant challenge for orthopaedic surgeons. Endothelial progenitor cells (EPCs) have successfully promoted the repair of acute defects in animal models; however, the ability of EPCs to induce the repair of chronic nonhealing defects, such as those often encountered clinically, has not been investigated. Therefore, the purpose of this study was to investigate the ability of EPCs delivered in delayed fashion to induce the repair of nonhealing defects in a clinically relevant model. In order to simulate delayed treatment, 5 mm segmental defects in Fischer 344 rat femora were treated with bone marrow-derived EPCs on a Gelfoam scaffold at 3 weeks post creation of the defect. At ten weeks posttreatment, 100% of EPC-treated defects achieved union, whereas complete union was only achieved in 37.5% of defects treated with Gelfoam alone. Furthermore, significant increases in ultimate torque (p = 0.022) and torsional stiffness (p = 0.003) were found in EPC-treated defects compared to controls. Critically, no differences in outcomes were observed between acute and delayed EPC treatments. These results suggest that EPCs can enhance bone healing when applied in an acute or delayed fashion and that their use may represent a clinically translatable therapy for bone healing in humans.
Collapse
|
170
|
Diaper R, Wong E, Metcalfe SA. The implications of biologic therapy for elective foot and ankle surgery in patients with rheumatoid arthritis. Foot (Edinb) 2017; 30:53-58. [PMID: 28262590 DOI: 10.1016/j.foot.2017.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 12/30/2016] [Accepted: 01/25/2017] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is one of a number of inflammatory arthropathies resulting in foot pain and deformity. Patients with this disease may require surgical intervention as part of their management. Many of these patients are now taking biologic agents which pose several risks to patients in the perioperative phase. The surgical team therefore need to be aware of these associated complications and how to manage these cases. AIM This paper aims to review the current literature about perioperative needs (foot and ankle surgery) associated with patients with rheumatoid arthritis receiving biologic therapy. MAIN FINDINGS The majority of the literature discusses the perioperative complications associated with patients on anti-TNFα therapy with few studies investigating the other biologics in common use. There is conflicting evidence as to the safety of continuing or stopping biologic drug therapy prior to orthopaedic procedures. The British Society for Rheumatology (BSR) have produced guidelines for the management of patients on anti-TNFα therapy or the biologic agent Tocilizumab. These recommendations suggest the risks of post-operative infection need to be balanced against the risk of a post-operative disease flare. In essence, it is suggested anti-TNFα therapy is stopped 3-5 times the half-life of the drug whilst Tocilizumab is stopped 4 weeks prior to surgery. CONCLUSION Good communication is needed between the surgical team and the local Rheumatology department managing the patient's disease in order to optimise perioperative care. Local pathways may vary from the BSR recommendations to determine the most suitable course of action with regards to continuing or stopping biologic therapy prior to foot and ankle surgery.
Collapse
Affiliation(s)
- Ross Diaper
- Podiatry Department, Turner Centre, St. James Hospital, Locksway Road, Portsmouth PO4 8LD, UK.
| | - Ernest Wong
- Department of Rheumatology, Portsmouth Hospitals Trust, Queen Alexandra Hospital, Portsmouth PO6 3LY, UK
| | - Stuart A Metcalfe
- Podiatry Department, Turner Centre, St. James Hospital, Locksway Road, Portsmouth PO4 8LD, UK
| |
Collapse
|
171
|
Abstract
Bone, despite its relatively inert appearance, is a tissue that is capable of adapting to its environment. Wolff’s law, first described in the 19th century, describes the ability of bone to change structure depending on the mechanical forces applied to it. The mechanostat model extended this principle and suggested that the amount of strain a bone detects depends on bone strength and the amount of muscle force applied to the bone. Experimental studies have found that low-magnitude, high-frequency mechanical loading is considered to be the most effective at increasing bone formation. The osteocyte is considered to be the master regulator of the bone response to mechanical loading. Deformation of bone matrix by mechanical loading is thought to result in interstitial fluid flow within the lacunar–canalicular system, which may activate osteocyte mechanosensors, leading to changes in osteocyte gene expression and ultimately increased bone formation and decreased bone resorption. However, repetitive strain applied to bone can result in microcracks, which may propagate and coalesce, and if not repaired predispose to catastrophic fracture. Osteocytes are a key component in this process, whereby apoptotic osteocytes in an area of microdamage promote targeted remodeling of the damaged bone. If fractures do occur, fracture repair can be divided into 2 types: primary and secondary healing. Secondary fracture repair is the most common and is a multistage process consisting of hematoma formation and acute inflammation, callus formation, and finally remodeling, whereby bone may return to its original form.
Collapse
|
172
|
Franco-Barrera MJ, Zavala-Cerna MG, Aguilar-Portillo G, Sánchez-Gomez DB, Torres-Bugarin O, Franco-Barrera MA, Roa-Encarnacion CM. Gorham-Stout Disease: a Clinical Case Report and Immunological Mechanisms in Bone Erosion. Clin Rev Allergy Immunol 2016; 52:125-132. [DOI: 10.1007/s12016-016-8594-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
173
|
Manfrini M, Mazzoni E, Barbanti-Brodano G, Nocini P, D'agostino A, Trombelli L, Tognon M. Osteoconductivity of Complex Biomaterials Assayed by Fluorescent-Engineered Osteoblast-like Cells. Cell Biochem Biophys 2016; 71:1509-15. [PMID: 25388843 DOI: 10.1007/s12013-014-0374-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Biomaterials employed for the bone regeneration can be assayed for specific features such as osteoconductivity and gene expression. In this study, the composite HA/collagen/chondroitin-sulfate biomaterial was investigated using an engineered human cell line, named Saos-eGFP. This cell line, a green fluorescent engineered human osteoblast-like cell, was employed as a cellular model for the in vitro study of biomaterial characteristics. The cytotoxicity was indirectly evaluated by fluorescence detection, osteoconductivity was assayed both by fluorescence and electron microscope analysis as well as cell morphology, whereas the RT-PCR technique was employed to assay gene expression. Saos-eGFP cells viability detection after 24 and 96 h of incubation showed that biomaterial enables the adhesion and proliferation of seeded cells as well as that of the plastic surface, the control. Fluorescence and scanning electron microscopy (SEM) analyses indicated that Saos-eGFP cells were homogeneously distributed on the HA granule surfaces, exhibiting cytoplasmic bridges, and were localized on the collagen-chondroitin sulfate extra-cellular matrix. An expression analysis of specific genes encoding for differentiation markers, showed that biomaterial assayed did not alter the osteogenic pathway of the Saos-eGFP cell line. Our assays confirm the cytocompatibility of this biomaterial, suggesting an osteoconductive capacity mediated by its chemical contents. We showed that the Saos-eGFP cellular model is suitable for in vitro biomaterial assays, and more specifically for assessing osteoconductivity. This result suggests that the cytocompatibility and osteoconductive features of the biomaterial assayed as bone substitute, could have a positive downstream effect on implant osteo-integration.
Collapse
Affiliation(s)
- Marco Manfrini
- Department of Morphology, Surgery and Experimental Medicine, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Morphology, Surgery and Experimental Medicine, School of Medicine, University of Ferrara, Ferrara, Italy
| | | | | | - Antonio D'agostino
- Department of Surgery, School of Medicine, University of Verona, Verona, Italy
| | - Leonardo Trombelli
- Research Centre for the Study of Periodontal and Peri-Implant Diseases, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Morphology, Surgery and Experimental Medicine, School of Medicine, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
174
|
Xiao W, Hu Z, Li T, Li J. Bone fracture healing is delayed in splenectomic rats. Life Sci 2016; 173:55-61. [PMID: 27956350 DOI: 10.1016/j.lfs.2016.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/02/2016] [Accepted: 12/09/2016] [Indexed: 12/28/2022]
Abstract
AIMS Splenectomy is sometimes required in bone fracture patients. The present study aims to investigate the effect of splenectomy on fracture healing in rats. MAIN METHODS Rats underwent osteotomy were subjected to splenectomy. The effects of splenectomy were evaluated at day 0, 3, 15 and 30 post-fracture. Double immunofluorescence staining was used to examine the expression of macrophages marker F4/80 and CD11b. H&E staining was used to examine the histological changes in fracture sites. Western blotting was used to detect protein expression of osteoprotegerin (OPG), the ligand for receptor activator of NF-κB (RANKL) and collagens in the fracture site. Activity of alkaline phosphatase (ALP) in the serum was determined using a biochemical kit. Serum levels of osteocalcin and inflammatory cytokines were determined using ELISA kits. Real-time PCR was used to detect mRNA expression of ALP and osteocalcin in the fracture site. KEY FINDINGS Results showed that the recruitment of macrophages and the production of inflammatory cytokines were inhibited in the fracture rats underwent splenectomy. Importantly, histological examination showed that fracture healing was delayed in splenectomized rats. In addition, the protein expression of OPG and RANKL in the fracture site was diminished, the activity of ALP and the level of osteocalcin in the serum and their mRNA expression in the fracture site were reduced, and the protein expression of type I collage a1 and type II collage a1 was inhibited in fracture rats underwent splenectomy compared with that in rats without splenectomy. SIGNIFICANCE Our findings indicate that splenectomy delays fracture healing.
Collapse
Affiliation(s)
- Wan'an Xiao
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang 110036, People's Republic of China
| | - Zhenyu Hu
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang 110036, People's Republic of China
| | - Tianwei Li
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang 110036, People's Republic of China
| | - Jianjun Li
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang 110036, People's Republic of China.
| |
Collapse
|
175
|
Lin JP, Shi ZJ, Shen NJ, Wang J, Li ZM, Xiao J. Serum N-terminal telopeptide of type I collagen as an early marker of fracture nonunion in rabbits. Exp Ther Med 2016; 12:3595-3601. [PMID: 28105092 PMCID: PMC5228403 DOI: 10.3892/etm.2016.3839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/08/2016] [Indexed: 11/17/2022] Open
Abstract
The aim of the present study was to establish an experimental animal model of fracture nonunion, and to investigate the changes in serum biomarker concentrations in fracture nonunion. A total of 20 purebred New Zealand rabbits were divided into two group: A bone defect group and a bone fracture group. In the bone defect group, a 15-mm section of bone (including the periosteum) was removed from the mid-radius, and the medullary cavities were closed with bone wax. In the bone fracture group, the mid-radius was fractured. X-rays were taken and blood samples were collected preoperatively and at 2, 3, 4, 5, 6, 7, 8, 10 and 12 weeks after the surgical procedure. The serum concentrations of osteocalcin (OC) and bone-specific alkaline phosphatase (BSAP) served as markers of bone formation, and those of C-terminal telopeptide of type I collagen (CTX), N-terminal telopeptide of type I collagen (NTX) and tartrate-resistant acid phosphatase 5b (TRACP 5b) served as markers of bone resorption. The concentration levels of the markers were measured using a biotin double-antibody sandwich enzyme-linked immunosorbent assay. In the bone defect group, bone callus was observed on X-ray at 2 weeks in three rabbits and the bone calluses stabilized at 5 weeks; however, none of the bones had healed at 8 weeks. In the bone fracture group, the fracture line was distorted at 2 weeks and bone calluses formed at 6-8 weeks. In the bone defect group, the serum BSAP and TRACP 5b concentrations increased following the surgical procedure, peaked at 4 weeks, began to decrease at 5 weeks and stabilized after 6 weeks. The serum OC concentrations did not change significantly following the surgical procedure. The serum CTX concentrations fluctuated during the first 4 weeks, peaked at 5 weeks, then decreased and stabilized after 6 weeks. The serum NTX concentrations fluctuated during the first 4 weeks, were significantly lower at 5 weeks compared with the other time points and stabilized after 6 weeks. These results suggested that a bone nonunion model can be established in New Zealand rabbits by resecting a 15-mm section of bone from the mid-radius prior to bone wax blocking. Measurement of the serum BSAP, CTX, NTX, and TRACP 5b concentrations may be useful for the early detection of bone nonunion. The serum NTX concentrations changed significantly in rabbits with bone nonunion. Further studies are required in order to determine the feasibility of using serum NTX concentrations for the early diagnosis of bone nonunion.
Collapse
Affiliation(s)
- Jian-Ping Lin
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Department of Orthopedic Surgery, Hainan Provincial People's Hospital, Haikou, Hainan 570311, P.R. China
| | - Zhan-Jun Shi
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ning-Jiang Shen
- Department of Orthopedic Surgery, Hainan Provincial People's Hospital, Haikou, Hainan 570311, P.R. China
| | - Jian Wang
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zao-Min Li
- Department of Orthopedic Surgery, Hainan Provincial People's Hospital, Haikou, Hainan 570311, P.R. China
| | - Jun Xiao
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
176
|
Lin HN, Cottrell J, O'Connor JP. Variation in lipid mediator and cytokine levels during mouse femur fracture healing. J Orthop Res 2016; 34:1883-1893. [PMID: 26919197 DOI: 10.1002/jor.23213] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/23/2016] [Indexed: 02/04/2023]
Abstract
Fracture healing is regulated by a variety of inflammatory mediators and growth factors which act over time to regenerate the injured tissue. This study used a mouse femur fracture model to quantify the temporal expression pattern of lipid mediators, cytokines, and related mRNAs during healing. Cyclooxygenase (COX-1 and COX-2) and 5-lipoxygenase (5-LO) derived lipid mediators, cytokines, and mRNA levels were quantified using mass spectrometry (LC-MS/MS), bead-based multiplex assays (xMAP), and quantitative PCR of cDNA (RTqPCR), respectively. Our analysis found that, the early inflammatory response (between 0 and 4 days after fracture) in the mouse femur fracture model coincided with elevated levels of COX-derived lipid mediators and inflammatory cytokines but with decreased levels of 5-LO-derived lipid mediators. Further, the COX-derived lipid mediators remained elevated for at least 7 days after fracture, suggesting that the COX-derived lipid mediators have additional functions during later phases of the fracture healing response. Differences were also found between mRNA levels and corresponding cytokines and lipid mediator levels, supporting a role for post-transcriptional regulation of gene expression. The temporal changes in fracture callus lipid mediator levels and inflammatory cytokines support a general positive role for inflammatory cytokines and COX-derived lipid mediators on fracture healing and a general negative role for 5-lipoxygenase derived lipid mediators during the initial stages of repair. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1883-1893, 2016.
Collapse
Affiliation(s)
- Hsuan-Ni Lin
- Department of Biochemistry and Molecular Biology, Rutgers, the State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, New Jersey 07103.,Department of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, 185 South Orange Avenue, Newark, New Jersey 07103
| | - Jessica Cottrell
- Department of Biochemistry and Molecular Biology, Rutgers, the State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, New Jersey 07103.,Department of Biological Sciences, Seton Hall University, 400 South Orange Avenue, South Orange, New Jersey 07079
| | - J Patrick O'Connor
- Department of Biochemistry and Molecular Biology, Rutgers, the State University of New Jersey, New Jersey Medical School, 185 South Orange Avenue, Newark, New Jersey 07103.,Department of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, 185 South Orange Avenue, Newark, New Jersey 07103
| |
Collapse
|
177
|
El-Jawhari JJ, Jones E, Giannoudis PV. The roles of immune cells in bone healing; what we know, do not know and future perspectives. Injury 2016; 47:2399-2406. [PMID: 27809990 DOI: 10.1016/j.injury.2016.10.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Key events occurring during the bone healing include well-orchestrated and complex interactions between immune cells, multipotential stromal cells (MSCs), osteoblasts and osteoclasts. Through three overlapping phases of this physiological process, innate and adaptive immune cells, cytokines and chemokines have a significant role to play. The aim of the escalating immune response is to achieve an osseous healing in the shortest time and with the least complications facilitating the restoration of function. The uninterrupted progression of these biological events in conjunction with a favourable mechanical environment (stable fracture fixation) remains the hallmark of successful fracture healing. When failure occurs, either the biological environment or the mechanical one could have been disrupted. Not infrequently both may be compromised. Consequently, regenerative treatments involving the use of bone autograft, allograft or synthetic matrices supplemented with MSCs are increasingly used. A better understanding of the bone biology and osteoimmunology can help to improve these evolving cell-therapy based strategies. Herein, an up to date status of the role of immune cells during the different phases of bone healing is presented. Additionally, the known and yet to know events about immune cell interactions with MSCs and osteoblasts and osteoclasts and the therapeutic implications are being discussed.
Collapse
Affiliation(s)
- Jehan J El-Jawhari
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St. James Hospital, University of Leeds, UK; NIHR Biomedical Research Unit, Chapel Allerton Hospital, University of Leeds, UK; Clinical Pathology Department, Faculty of Medicine, Mansoura University, Egypt
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St. James Hospital, University of Leeds, UK; NIHR Biomedical Research Unit, Chapel Allerton Hospital, University of Leeds, UK
| | - Peter V Giannoudis
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St. James Hospital, University of Leeds, UK; NIHR Biomedical Research Unit, Chapel Allerton Hospital, University of Leeds, UK.
| |
Collapse
|
178
|
Breast cancer cells and bone marrow mesenchymal stromal cells: a regulated modulation of the breast tumor in the context of immune response. Inflamm Res 2016; 66:129-139. [PMID: 27783097 DOI: 10.1007/s00011-016-1000-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE The role of direct cell-cell interactions mediating selective bone metastasis by breast cancer cells (BCCs) niche is still mostly unknown. MATERIALS AND METHODS Conditioned medium and direct cell-cell contacts experiments were used to investigate the effect of bone marrow-derived mesenchymal stromal cells (MSCs), osteoprogenitor-like cells (MG-63) and osteosarcoma cells (SaOS-2) on luminal-like (MCF-7) and basal-like (MDA-MB-231) BCCs flow cytometry was used to assess the purity of isolated BCCs and osteoblasts. Expression of osteoblastic markers was investigated by semi-quantitative RT-PCR. RANKL and OPG levels were measured by ELISA. RESULTS Conditioned medium from MSCs and osteoblasts induced the expression of osteoblastic markers in BCCs. While co-culture assays with SaOS-2 increased the expression of osteoblastic markers in MCF-7 cells, SaOS-2 cell conditioned medium increased the expression of RANKL, PTHrP, VEGF and NOGGIN in MCF-7 cells. Co-cultures with either MG-63 cells or MSCs induced OPG and MMP-2 in both tumor cell lines. Interestingly, conditioned medium from co-cultures of MSCs and MDA-MB-231 cells significantly decreased the proliferation of activated T lymphocytes which was reversed by addition of anti-OPG antibodies to the co-cultures. CONCLUSION Our data suggest that MSCs strongly contribute to the adaptation and invasiveness of breast cancer cells in skeletal tissues.
Collapse
|
179
|
Wang X, Friis TE, Masci PP, Crawford RW, Liao W, Xiao Y. Alteration of blood clot structures by interleukin-1 beta in association with bone defects healing. Sci Rep 2016; 6:35645. [PMID: 27767056 PMCID: PMC5073366 DOI: 10.1038/srep35645] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/30/2016] [Indexed: 12/18/2022] Open
Abstract
The quality of hematomas are crucial for successful early bone defect healing, as the structure of fibrin clots can significantly influence the infiltration of cells, necessary for bone regeneration, from adjacent tissues into the fibrin network. This study investigated if there were structural differences between hematomas from normal and delayed healing bone defects and whether such differences were linked to changes in the expression of IL-1β. Using a bone defect model in rats, we found that the hematomas in the delayed healing model had thinner fibers and denser clot structures. Moreover, IL-1β protein levels were significantly higher in the delayed healing hematomas. The effects of IL-1β on the structural properties of human whole blood clots were evaluated by thrombelastograph (TEG), scanning electronic microscopy (SEM), compressive study, and thrombolytic assays. S-nitrosoglutathione (GSNO) was applied to modulate de novo hematoma structure and the impact on bone healing was evaluated in the delayed healing model. We found that GSNO produced more porous hematomas with thicker fibers and resulted in significantly enhanced bone healing. This study demonstrated that IL-1β and GSNO had opposing effects on clot architecture, the structure of which plays a pivotal role in early bone healing.
Collapse
Affiliation(s)
- Xin Wang
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 4059 Queensland, Australia
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou, China
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, 4059 Queensland, Australia
- Translational Research Institute, School of Medicine, The University of Queensland, Brisbane, 4102 Queensland, Australia
| | - Thor E. Friis
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 4059 Queensland, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, 4059 Queensland, Australia
| | - Paul P. Masci
- Translational Research Institute, School of Medicine, The University of Queensland, Brisbane, 4102 Queensland, Australia
| | - Ross W. Crawford
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 4059 Queensland, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, 4059 Queensland, Australia
| | - Wenbo Liao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou, China
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 4059 Queensland, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, 4059 Queensland, Australia
| |
Collapse
|
180
|
Abstract
Background and purpose - Studies of fracture healing have mainly dealt with shaft fractures, both experimentally and clinically. In contrast, most patients have metaphyseal fractures. There is an increasing awareness that metaphyseal fractures heal partly through mechanisms specific to cancellous bone. Several new models for the study of cancellous bone healing have recently been presented. This review summarizes our current knowledge of cancellous fracture healing. Methods - We performed a review of the literature after doing a systematic literature search. Results - Cancellous bone appears to heal mainly via direct, membranous bone formation that occurs freely in the marrow, probably mostly arising from local stem cells. This mechanism appears to be specific for cancellous bone, and could be named inter-trabecular bone formation. This kind of bone formation is spatially restricted and does not extend more than a few mm outside the injured region. Usually no cartilage is seen, although external callus and cartilage formation can be induced in meta-physeal fractures by mechanical instability. Inter-trabecular bone formation seems to be less sensitive to anti-inflammatory treatment than shaft fractures. Interpretation - The unique characteristics of inter-trabecular bone formation in metaphyseal fractures can lead to differences from shaft healing regarding the effects of age, loading, or drug treatment. This casts doubt on generalizations about fracture healing based solely on shaft fracture models.
Collapse
|
181
|
Villafan-Bernal JR, Franco-De La Torre L, Sandoval-Rodriguez AS, Armendariz-Borunda J, Alcala-Zermeno JL, Cruz-Ramos JA, Lopez-Armas G, Ramirez-Bastidas BE, González-Enríquez GV, Collazo-Guzman EA, Martinez-Portilla RJ, Sánchez-Enríquez S. Molecular profiling of a simple rat model of open tibial fractures with hematoma and periosteum disruption. Exp Ther Med 2016; 12:3261-3267. [PMID: 27882147 PMCID: PMC5103776 DOI: 10.3892/etm.2016.3758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 07/26/2016] [Indexed: 01/22/2023] Open
Abstract
Bone fractures are a worldwide public health concern. Therefore, improving understanding of the bone healing process at a molecular level, which could lead to the discovery of potential therapeutic targets, is important. In the present study, a model of open tibial fractures with hematoma disruption, periosteal rupture and internal fixation in 6-month-old male Wistar rats was established, in order to identify expression patterns of key genes and their protein products throughout the bone healing process. A tibial shaft fracture was produced using the three-point bending technique, the hematoma was drained through a 4-mm incision on the medial aspect of the tibia and the fracture stabilized by inserting a needle into the medullary canal. Radiographs confirmed that the induced fractures were diaphyseal and this model was highly reproducible (kappa inter-rater reliability, 0.82). Rats were sacrificed 5, 14, 21, 28 and 35 days post-fracture to obtain samples for histological, immunohistochemical and molecular analysis. Expression of interleukin-1β (Il-1β), transforming growth factor-β2 (Tgf-β2), bone morphogenetic protein-6 (Bmp-6), bone morphogenetic protein-7 (Bmp-7) and bone γ-carboxyglutamic acid-containing protein (Bglap) genes was determined by reverse transcription quantitative polymerase chain reaction and protein expression was evaluated by immunohistochemistry, while histological examination allowed characterization of the bone repair process. Il-1β showed a biphasic expression, peaking 5 and 28 days post-fracture. Expression of Tgf-β2, Bmp-6 and Bmp-7 was restricted to the period 21 days post-fracture. Bglap expression increased gradually, peaking 21 days post-fracture, although it was expressed in all evaluated stages. Protein expression corresponded with the increased expression of their corresponding genes. In conclusion, a clear and well-defined expression pattern of the evaluated genes and proteins was observed, where their maximal expression correlated with their known participation in each stage of the bone healing process.
Collapse
Affiliation(s)
- Jose Rafael Villafan-Bernal
- Young Scientist and Cathedras Department, National Council of Science and Technology (CONACYT), Mexico City 03940, Mexico; Molecular Biology and Genomics Department, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico; Surgery Department, Health Science University Center, Autonomous University of Aguascalientes, Aguascalientes, Aguascalientes 20131, Mexico
| | - Lorenzo Franco-De La Torre
- Molecular Biology and Genomics Department, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico; Health Sciences Department, Los Altos University Center at Tepatitlan, University of Guadalajara, Guadalajara, Jalisco 47620, Mexico
| | - Ana Soledad Sandoval-Rodriguez
- Institute of Molecular Biology in Medicine and Gene Therapy, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Juan Armendariz-Borunda
- Institute of Molecular Biology in Medicine and Gene Therapy, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Juan Luis Alcala-Zermeno
- Molecular Biology and Genomics Department, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | | | - Gabriela Lopez-Armas
- Technical and Industrial Teaching Center (CETI) Colomos, Guadalajara, Jalisco 44638, Mexico
| | - Blanca Estela Ramirez-Bastidas
- Molecular Biology and Genomics Department, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico; Chronic and Degenerative Disease Institute, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Gracia Viviana González-Enríquez
- Methodological and Instrumental Disciplines Department, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| | - Emerson Armando Collazo-Guzman
- Surgery Department, Health Science University Center, Autonomous University of Aguascalientes, Aguascalientes, Aguascalientes 20131, Mexico
| | - Raigam Jafet Martinez-Portilla
- Surgery Department, Health Science University Center, Autonomous University of Aguascalientes, Aguascalientes, Aguascalientes 20131, Mexico
| | - Sergio Sánchez-Enríquez
- Molecular Biology and Genomics Department, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco 44340, Mexico
| |
Collapse
|
182
|
Haubruck P, Kammerer A, Korff S, Apitz P, Xiao K, Büchler A, Biglari B, Zimmermann G, Daniel V, Schmidmaier G, Moghaddam A. The treatment of nonunions with application of BMP-7 increases the expression pattern for angiogenic and inflammable cytokines: a matched pair analysis. J Inflamm Res 2016; 9:155-165. [PMID: 27703392 PMCID: PMC5036623 DOI: 10.2147/jir.s110621] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The local application of bone morphogenetic protein-7 (BMP-7) in combination with the transplantation of autologous bone graft improves the outcome in nonunion treatment; however, the specific reasons remain unclear. In this study, we sought to determine if the local application of BMP-7 contributes to improved bone regeneration in nonunion therapy by modulation of the angiogenic and inflammable cytokine expression patterns of the early inflammation response. Therefore, we utilized the analysis of serological cytokine expression patterns. As a matched pair analysis, best-fitting patients who were treated with transplantation of autologous bone graft (G1, n=10) were compared with patients who were treated with additional application of BMP-7 (G2, n=10). The changes in the cytokine expression patterns were monitored and correlated to clinical data of bone healing. Significant differences in angiogenesis potential (vascular endothelial growth factor [VEGF] serum levels) could be found in the first days after surgery (P<0.05). Furthermore, the increase and absolute amount of VEGF levels in the BMP-7 group were considerably higher than in the control group during the first 2 weeks after surgery. The expression pattern of inflammable cytokines showed noticeable differences in the time point of significant elevated levels, in particular, inflammable cytokines showed an earlier peak in G2. Furthermore, interleukin-6 was significantly elevated within the first week only, comparing G2 to G1 (P<0.05). Our findings indicate that BMP-7 induces an early and more intense expression of VEGF via a direct and postulated indirect pathway, thereby providing a favorable environment for bone healing. Moreover, application of BMP-7 leads to an earlier expression of known proinflammatory cytokines. The results of this study show that application of BMP-7 leads to costimulatory effect on both angiogenic and inflammable cytokine expression patterns that may serve as a possible stimulus for bone regeneration.
Collapse
Affiliation(s)
- Patrick Haubruck
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Andreas Kammerer
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Sebastian Korff
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Philipp Apitz
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Kai Xiao
- Department of Orthopedics Wuhan, Wuhan Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Axel Büchler
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Bahram Biglari
- Department of Paraplegiology, Berufsgenossenschaftliche Unfallklinik Ludwigshafen, Ludwigshafen
| | - Gerald Zimmermann
- Department for Trauma Surgery, Theresienkrankenhaus und St. Hedwigs-Klinik GmbH, Mannheim
| | - Volker Daniel
- Department of Transplantation Immunology, Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Gerhard Schmidmaier
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| | - Arash Moghaddam
- Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, HTRG - Heidelberg Trauma Research Group, Heidelberg, Germany
| |
Collapse
|
183
|
Cytokines TNF-α, IL-6, IL-17F, and IL-4 Differentially Affect Osteogenic Differentiation of Human Adipose Stem Cells. Stem Cells Int 2016; 2016:1318256. [PMID: 27667999 PMCID: PMC5030432 DOI: 10.1155/2016/1318256] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/09/2016] [Accepted: 08/16/2016] [Indexed: 12/16/2022] Open
Abstract
During the initial stages of bone repair, proinflammatory cytokines are released within the injury site, quickly followed by a shift to anti-inflammatory cytokines. The effect of pro- and anti-inflammatory cytokines on osteogenic differentiation of mesenchymal stem cells is controversial. Here, we investigated the effect of the proinflammatory cytokines TNF-α, IL-6, IL-8, and IL-17F and the anti-inflammatory cytokine IL-4 on proliferation and osteogenic differentiation of human adipose stem cells (hASCs). hASCs were treated with TNF-α, IL-6, IL-8, IL-17F, or IL-4 (10 ng/mL) for 72 h mimicking bone repair. TNF-α reduced collagen type I gene expression but increased hASC proliferation and ALP activity. IL-6 also strongly enhanced ALP activity (18-fold), as well as bone nodule formation by hASCs. IL-8 did not affect proliferation or osteogenic gene expression but reduced bone nodule formation. IL-17F decreased hASC proliferation but enhanced ALP activity. IL-4 enhanced osteocalcin gene expression and ALP activity but reduced RUNX2 gene expression and bone nodule formation. In conclusion, all cytokines studied have both enhancing and reducing effects on osteogenic differentiation of hASCs, even when applied for 72 h only. Some cytokines, specifically IL-6, may be suitable to induce osteogenic differentiation of mesenchymal stem cells as a strategy for enhancing bone repair.
Collapse
|
184
|
Bian H, Lin JZ, Li C, Farmer SR. Myocardin-related transcription factor A (MRTFA) regulates the fate of bone marrow mesenchymal stem cells and its absence in mice leads to osteopenia. Mol Metab 2016; 5:970-979. [PMID: 27689009 PMCID: PMC5034694 DOI: 10.1016/j.molmet.2016.08.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/17/2016] [Accepted: 08/19/2016] [Indexed: 01/02/2023] Open
Abstract
Objective Arising from common progenitors in the bone marrow, adipogenesis and osteogenesis are closely associated yet mutually exclusive during bone marrow mesenchymal stem cell (BMSC) development. Previous studies have shown that morphological changes can affect the early commitment of pluripotent BMSCs to the adipose versus osteoblastic lineage via modulation of RhoA activity. The RhoA pathway regulates actin polymerization to promote the incorporation of globular actin (G-actin) into filamentous actin (F-actin). In doing so, myocardin-related transcription factors (MRTFs) dissociate from bound G-actin and enter the nucleus to co-activate serum response factor (SRF) target gene expression. In this study, we investigated whether MRTFA/SRF is acting downstream of the RhoA pathway to regulate BMSC commitment in mice. Methods The effects of knocking out MRTFA on skeletal homeostasis was studied in MRTFA KO mice using micro-CT, QPCR and western blot assays. To determine how MRTFA affects the mechanisms regulating BMSC fate decisions, primary bone marrow stromal cells from WT and MRTFA KO mice as well as C3H10T1/2 cell lines were analyzed in vitro. Results Global MRTFA KO mice have lower whole body weight, shorter femoral and tibial lengths as well as significantly decreased bone mass in their femurs. BMSCs isolated from the KO mice show increased adipogenesis and reduced osteogenesis when compared to WT littermates. KO mice, particularly females, develop osteopenia with age, and this was enhanced by a high fat diet. Over-expression of MRTFA or SRF enhances osteogenesis in CH310T1/2 cell lines. Sca1+, CD45− cells from KO marrow express lower amounts of smooth muscle actin (SMA) and TAZ/YAP target genes compared to WT counterparts. Conclusion This study identified MRTFA as a novel regulator of skeletal homeostasis by regulating the balance between adipogenic and osteogenic differentiation of BMSCs. We propose that MRTFA promotes the osteogenic activity of TAZ/YAP by maintaining SMA production in BMSCs.
Collapse
Affiliation(s)
- Hejiao Bian
- Department of Biochemistry, Boston University School of Medicine, 72 East Concord Street, K606A, Boston, MA 02118, USA
| | - Jean Z Lin
- Department of Biochemistry, Boston University School of Medicine, 72 East Concord Street, K606A, Boston, MA 02118, USA
| | - Chendi Li
- Department of Biochemistry, Boston University School of Medicine, 72 East Concord Street, K606A, Boston, MA 02118, USA
| | - Stephen R Farmer
- Department of Biochemistry, Boston University School of Medicine, 72 East Concord Street, K606A, Boston, MA 02118, USA.
| |
Collapse
|
185
|
Carpio LR, Bradley EW, McGee-Lawrence ME, Weivoda MM, Poston DD, Dudakovic A, Xu M, Tchkonia T, Kirkland JL, van Wijnen AJ, Oursler MJ, Westendorf JJ. Histone deacetylase 3 supports endochondral bone formation by controlling cytokine signaling and matrix remodeling. Sci Signal 2016; 9:ra79. [PMID: 27507649 PMCID: PMC5409103 DOI: 10.1126/scisignal.aaf3273] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Histone deacetylase (HDAC) inhibitors are efficacious epigenetic-based therapies for some cancers and neurological disorders; however, each of these drugs inhibits multiple HDACs and has detrimental effects on the skeleton. To better understand how HDAC inhibitors affect endochondral bone formation, we conditionally deleted one of their targets, Hdac3, pre- and postnatally in type II collagen α1 (Col2α1)-expressing chondrocytes. Embryonic deletion was lethal, but postnatal deletion of Hdac3 delayed secondary ossification center formation, altered maturation of growth plate chondrocytes, and increased osteoclast activity in the primary spongiosa. HDAC3-deficient chondrocytes exhibited increased expression of cytokine and matrix-degrading genes (Il-6, Mmp3, Mmp13, and Saa3) and a reduced abundance of genes related to extracellular matrix production, bone development, and ossification (Acan, Col2a1, Ihh, and Col10a1). Histone acetylation increased at and near genes that had increased expression. The acetylation and activation of nuclear factor κB (NF-κB) were also increased in HDAC3-deficient chondrocytes. Increased cytokine signaling promoted autocrine activation of Janus kinase (JAK)-signal transducer and activator of transcription (STAT) and NF-κB pathways to suppress chondrocyte maturation, as well as paracrine activation of osteoclasts and bone resorption. Blockade of interleukin-6 (IL-6)-JAK-STAT signaling, NF-κB signaling, and bromodomain extraterminal proteins, which recognize acetylated lysines and promote transcriptional elongation, significantly reduced Il-6 and Mmp13 expression in HDAC3-deficient chondrocytes and secondary activation in osteoclasts. The JAK inhibitor ruxolitinib also reduced osteoclast activity in Hdac3 conditional knockout mice. Thus, HDAC3 controls the temporal and spatial expression of tissue-remodeling genes and inflammatory responses in chondrocytes to ensure proper endochondral ossification during development.
Collapse
Affiliation(s)
- Lomeli R Carpio
- Mayo Graduate School, Mayo Clinic, Rochester, MN 55905, USA. Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA. Institute of Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, USA
| | - Megan M Weivoda
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel D Poston
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA. Creighton University, Omaha, NE 68102, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Ming Xu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre J van Wijnen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA. Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Merry Jo Oursler
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA. Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jennifer J Westendorf
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA. Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
186
|
Chrcanovic BR, Kisch J, Albrektsson T, Wennerberg A. Factors Influencing Early Dental Implant Failures. J Dent Res 2016; 95:995-1002. [PMID: 27146701 DOI: 10.1177/0022034516646098] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The purpose of the present study was to assess the influence of local and systemic factors on the occurrence of dental implant failures up to the second-stage surgery (abutment connection). This retrospective study is based on 2,670 patients who received 10,096 implants and were consecutively treated with implant-supported prostheses between 1980 and 2014 at 1 specialist clinic. Several anatomic-, patient-, health-, and implant-related factors were collected. Descriptive statistics were used to describe the patients and implants. Univariate and multivariate logistic regression models were used at the patient level as well as the implant level to evaluate the effect of explanatory variables on the failure of implants up to abutment connection. A generalized estimating equation method was used for the implant-level analysis to account for the fact that repeated observations (several implants) were available for a single patient. Overall, 642 implants (6.36%) failed, of which 176 (1.74%) in 139 patients were lost up to second-stage surgery. The distribution of implants in sites of different bone quantities and qualities was quite similar between implants lost up to and after abutment connection. Smoking and the intake of antidepressants were the statistically significant predictors in the multivariate model (ClinicalTrials.gov NCT02369562).
Collapse
Affiliation(s)
- B R Chrcanovic
- Department of Prosthodontics, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - J Kisch
- Clinic for Prosthodontics, Centre of Dental Specialist Care, Malmö, Sweden
| | - T Albrektsson
- Department of Prosthodontics, Faculty of Odontology, Malmö University, Malmö, Sweden Department of Biomaterials, Göteborg University, Göteborg, Sweden
| | - A Wennerberg
- Department of Prosthodontics, Faculty of Odontology, Malmö University, Malmö, Sweden
| |
Collapse
|
187
|
Pigatto Mitihiro D, de Paula Ramos S, Corazza Montero J, Alves Campos A, de Oliveira Toginho Filho D, Dezan Garbelini CC. Effects of near-infrared LED therapy on experimental tooth replantation in rats. Dent Traumatol 2016; 33:32-37. [PMID: 27472656 DOI: 10.1111/edt.12301] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND/AIM Dental avulsion damages periodontal tissues and may induce dentoalveolar ankylosis and root resorption after replantation. The aim of this work was to evaluate the effects of light-emitting diode (LED) therapy after tooth replantation. MATERIAL AND METHODS The upper right incisors of 36 male Wistar rats were extracted and replanted after 15 min. The control group (Co, n = 18) was submitted to replantation and the LED group (n = 18) was submitted to replantation and LED therapy (940 nm, 4 J cm-2 ) for three consecutive days (0, 24, and 48 h). Six animals from each group were euthanized at 7, 14, and 21 days for histological analysis. The upper left incisors were used as a negative control. Data were compared using the Fisher exact test, considering P < 0.05. RESULTS After 7 days, the Co (73.3%) and LED (72.2%) groups presented areas of periodontal ligament necrosis. However, periodontal ligament necrosis was more frequent in the LED group at 14 (70%, P < 0.05) and 21 days (43.2%, P < 0.05) in relation to the Co group (33 and 21.6%). Inflammatory cell infiltration was more frequent in the Co group at 7 days (100%) than the LED group (24.4%, P < 0.05), but increased in the LED group after 21 days (83.3%, P < 0.05). Bone necrosis was more frequent in the LED group after 14 (79%, P < 0.05) and 21 days (60%, P < 0.005) than in the Co group (45% and 20%). Bone repair was evident in the Co group at 14 days. All of the replanted teeth had pulp necrosis. CONCLUSION The results suggest that the application of LED therapy up to 48 h after tooth replantation may delay periodontal ligament repair.
Collapse
Affiliation(s)
- Débora Pigatto Mitihiro
- Department of Oral Medicine e Pediatric Dentistry, State University of Londrina, Londrina, Brasil
| | | | - Janaína Corazza Montero
- Department of Oral Medicine e Pediatric Dentistry, State University of Londrina, Londrina, Brasil
| | | | | | | |
Collapse
|
188
|
Interleukin 6 and/or Interleukin 17A Modulate the OPG/RANKL System of MC3T3-E1 Murine Osteoblast Cell Line. Int J Pept Res Ther 2016. [DOI: 10.1007/s10989-016-9529-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
189
|
Pretell-Mazzini J, Subhawong T, Hernandez VH, Campo R. HIV and Orthopaedics: Musculoskeletal Manifestations and Outcomes. J Bone Joint Surg Am 2016; 98:775-86. [PMID: 27147691 DOI: 10.2106/jbjs.15.00842] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
➤Advances in combined antiretroviral therapy (cART) in recent years have transformed HIV infection into a chronic disease when treatment is available, increasing a patient's life expectancy and the chances that orthopaedic surgeons will encounter such patients in their clinical practice.➤Musculoskeletal manifestations in patients with HIV infection are common and sometimes are the initial presentation of the disease. Knowledge about neoplasms and associated conditions affecting muscle, bones, and joints is essential for successful management.➤Since the advent of cART, total joint arthroplasty has been shown to be a safe procedure; however, perioperative infection is still a small risk in patients with uncontrolled viral loads or CD4 counts of <400 cells/mm(3).➤With regard to trauma surgery, the rates of early and late infection around implants, as well as union rates, are comparable with those in the HIV-negative population; however, there is an increased risk of pulmonary, renal, and infectious or septic complications in the polytrauma setting.➤Factors such as CD4 count, nutritional status, cART therapy, viral load count, and other comorbidities (hemophilia, infection among intravenous drug users, etc.) should be considered when treating these patients in order to optimize their clinical outcomes.
Collapse
Affiliation(s)
- Juan Pretell-Mazzini
- Departments of Orthopaedic Surgery (J.P.-M. and V.H.H.) and Radiology (T.S.), and Division of Infectious Diseases, Department of Internal Medicine (R.C.), University of Miami Miller School of Medicine, Miami, Florida
| | - Ty Subhawong
- Departments of Orthopaedic Surgery (J.P.-M. and V.H.H.) and Radiology (T.S.), and Division of Infectious Diseases, Department of Internal Medicine (R.C.), University of Miami Miller School of Medicine, Miami, Florida
| | - Victor H Hernandez
- Departments of Orthopaedic Surgery (J.P.-M. and V.H.H.) and Radiology (T.S.), and Division of Infectious Diseases, Department of Internal Medicine (R.C.), University of Miami Miller School of Medicine, Miami, Florida
| | - Rafael Campo
- Departments of Orthopaedic Surgery (J.P.-M. and V.H.H.) and Radiology (T.S.), and Division of Infectious Diseases, Department of Internal Medicine (R.C.), University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
190
|
Loi F, Córdova LA, Pajarinen J, Lin TH, Yao Z, Goodman SB. Inflammation, fracture and bone repair. Bone 2016; 86:119-30. [PMID: 26946132 PMCID: PMC4833637 DOI: 10.1016/j.bone.2016.02.020] [Citation(s) in RCA: 811] [Impact Index Per Article: 90.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/30/2015] [Accepted: 02/29/2016] [Indexed: 12/20/2022]
Abstract
The reconstitution of lost bone is a subject that is germane to many orthopedic conditions including fractures and non-unions, infection, inflammatory arthritis, osteoporosis, osteonecrosis, metabolic bone disease, tumors, and periprosthetic particle-associated osteolysis. In this regard, the processes of acute and chronic inflammation play an integral role. Acute inflammation is initiated by endogenous or exogenous adverse stimuli, and can become chronic in nature if not resolved by normal homeostatic mechanisms. Dysregulated inflammation leads to increased bone resorption and suppressed bone formation. Crosstalk among inflammatory cells (polymorphonuclear leukocytes and cells of the monocyte-macrophage-osteoclast lineage) and cells related to bone healing (cells of the mesenchymal stem cell-osteoblast lineage and vascular lineage) is essential to the formation, repair and remodeling of bone. In this review, the authors provide a comprehensive summary of the literature related to inflammation and bone repair. Special emphasis is placed on the underlying cellular and molecular mechanisms, and potential interventions that can favorably modulate the outcome of clinical conditions that involve bone repair.
Collapse
Affiliation(s)
- Florence Loi
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Luis A Córdova
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA; Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Chile, Sergio Livingstone Polhammer 943, Independencia, 8380000 Santiago, Chile.
| | - Jukka Pajarinen
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Tzu-hua Lin
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Zhenyu Yao
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Stuart B Goodman
- 300 Pasteur Drive, Edwards Building, Room R116, Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA; 300 Pasteur Drive, Edwards Building, Room R114, Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
191
|
Nencini S, Ivanusic JJ. The Physiology of Bone Pain. How Much Do We Really Know? Front Physiol 2016; 7:157. [PMID: 27199772 PMCID: PMC4844598 DOI: 10.3389/fphys.2016.00157] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/11/2016] [Indexed: 01/23/2023] Open
Abstract
Pain is associated with most bony pathologies. Clinical and experimental observations suggest that bone pain can be derived from noxious stimulation of the periosteum or bone marrow. Sensory neurons are known to innervate the periosteum and marrow cavity, and most of these have a morphology and molecular phenotype consistent with a role in nociception. However, little is known about the physiology of these neurons, and therefore information about mechanisms that generate and maintain bone pain is lacking. The periosteum has received greater attention relative to the bone marrow, reflecting the easier access of the periosteum for experimental assessment. With the electrophysiological preparations used, investigators have been able to record from single periosteal units in isolation, and there is a lot of information available about how they respond to different stimuli, including those that are noxious. In contrast, preparations used to study sensory neurons that innervate the bone marrow have been limited to recording multi-unit activity in whole nerves, and whilst they clearly report responses to noxious stimulation, it is not possible to define responses for single sensory neurons that innervate the bone marrow. There is only limited evidence that peripheral sensory neurons that innervate bone can be sensitized or that they can be activated by multiple stimulus types, and at present this only exists in part for periosteal units. In the central nervous system, it is clear that spinal dorsal horn neurons can be activated by noxious stimuli applied to bone. Some can be sensitized under pathological conditions and may contribute in part to secondary or referred pain associated with bony pathology. Activity related to stimulation of sensory nerves that innervate bone has also been reported in neurons of the spinoparabrachial pathway and the somatosensory cortices, both known for roles in coding information about pain. Whilst these provide some clues as to the way information about bone pain is centrally coded, they need to be expanded to further our understanding of other central territories involved. There is a lot more to learn about the physiology of peripheral sensory neurons that innervate bone and their central projections.
Collapse
Affiliation(s)
- Sara Nencini
- Department of Anatomy and Neuroscience, University of Melbourne Melbourne, VIC, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Neuroscience, University of Melbourne Melbourne, VIC, Australia
| |
Collapse
|
192
|
Guided bone regeneration is promoted by the molecular events in the membrane compartment. Biomaterials 2016; 84:167-183. [DOI: 10.1016/j.biomaterials.2016.01.034] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 01/18/2016] [Indexed: 11/18/2022]
|
193
|
Alves CJ, Neto E, Sousa DM, Leitão L, Vasconcelos DM, Ribeiro-Silva M, Alencastre IS, Lamghari M. Fracture pain-Traveling unknown pathways. Bone 2016; 85:107-14. [PMID: 26851411 DOI: 10.1016/j.bone.2016.01.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 12/14/2015] [Accepted: 01/08/2016] [Indexed: 12/15/2022]
Abstract
An increase of fracture incidence is expected for the next decades, mostly due to the undeniable increase of osteoporotic fractures, associated with the rapid population ageing. The rise in sports-related fractures affecting the young and active population also contributes to this increased fracture incidence, and further amplifies the economical burden of fractures. Fracture often results in severe pain, which is a primary symptom to be treated, not only to guarantee individual's wellbeing, but also because an efficient management of fracture pain is mandatory to ensure proper bone healing. Here, we review the available data on bone innervation and its response to fracture, and discuss putative mechanisms of fracture pain signaling. In addition, the common therapeutic approaches to treat fracture pain are discussed. Although there is still much to learn, research in fracture pain has allowed an initial insight into the mechanisms involved. During the inflammatory response to fracture, several mediators are released and will putatively activate and sensitize primary sensory neurons, in parallel, intense nerve sprouting that occurs in the fracture callus area is also suggested to be involved in pain signaling. The establishment of hyperalgesia and allodynia after fracture indicates the development of peripheral and central sensitization, still, the underlying mechanisms are largely unknown. A major concern during the treatment of fracture pain needs to be the preservation of proper bone healing. However, the most common therapeutic agents, NSAIDS and opiates, can cause significant side effects that include fracture repair impairment. The understanding of the mechanisms of fracture pain signaling will allow the development of mechanisms-based therapies to effectively and safely manage fracture pain.
Collapse
Affiliation(s)
- Cecília J Alves
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Rua Alfredo Allen, 208, 4150-180 Porto, Portugal
| | - Estrela Neto
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Rua Alfredo Allen, 208, 4150-180 Porto, Portugal; Faculdade de Medicina, Universidade do Porto (FMUP), Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Daniela M Sousa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Rua Alfredo Allen, 208, 4150-180 Porto, Portugal
| | - Luís Leitão
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Rua Alfredo Allen, 208, 4150-180 Porto, Portugal; Instituto Ciências Biomédicas Abel Salazar (ICBAS), Universidade de Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Daniel M Vasconcelos
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Rua Alfredo Allen, 208, 4150-180 Porto, Portugal; Instituto Ciências Biomédicas Abel Salazar (ICBAS), Universidade de Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Manuel Ribeiro-Silva
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Rua Alfredo Allen, 208, 4150-180 Porto, Portugal; Faculdade de Medicina, Universidade do Porto (FMUP), Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; Serviço de Ortopedia e Traumatologia, Centro Hospitalar São João, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Inês S Alencastre
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Rua Alfredo Allen, 208, 4150-180 Porto, Portugal
| | - Meriem Lamghari
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Rua Alfredo Allen, 208, 4150-180 Porto, Portugal; Instituto Ciências Biomédicas Abel Salazar (ICBAS), Universidade de Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
194
|
Waki T, Lee SY, Niikura T, Iwakura T, Dogaki Y, Okumachi E, Oe K, Kuroda R, Kurosaka M. Profiling microRNA expression during fracture healing. BMC Musculoskelet Disord 2016; 17:83. [PMID: 26879131 PMCID: PMC4754871 DOI: 10.1186/s12891-016-0931-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 02/06/2016] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The discovery of microRNA (miRNA) has revealed a novel type of regulatory control for gene expression. Increasing evidence suggests that miRNA regulates chondrocyte, osteoblast, and osteoclast differentiation and function, indicating miRNA as key regulators of bone formation, resorption, remodeling, and repair. We hypothesized that the functions of certain miRNAs and changes to their expression pattern may play crucial roles during the process of fracture healing. METHODS Standard healing fractures and unhealing fractures produced by periosteal cauterization at the fracture site were created in femurs of seventy rats, with half assigned to the standard healing fracture group and half assigned to the nonunion group. At post-fracture days 3, 7, 10, 14, 21, and 28, total RNA including miRNA was extracted from the newly generated tissue at the fracture site. Microarray analysis was performed with miRNA samples from each group on post-fracture day 14. For further analysis, we selected highly up-regulated five miRNAs in the standard healing fracture group from the microarray data. Real-time PCR was performed with miRNA samples at each time point above mentioned to compare the expression levels of the selected miRNAs between standard healing fractures and unhealing fractures and investigate their time-course changes. RESULTS Microarray and real-time polymerase chain reaction (PCR) analyses on day 14 revealed that five miRNAs, miR-140-3p, miR-140-5p, miR-181a-5p, miR-181d-5p, and miR-451a, were significantly highly expressed in standard healing fractures compared with unhealing fractures. Real-time PCR analysis further revealed that in standard healing fractures, the expression of all five of these miRNAs peaked on day 14 and declined thereafter. CONCLUSION Our results suggest that the five miRNAs identified using microarray and real-time PCR analyses may play important roles during fracture healing. These findings provide valuable information to further understand the molecular mechanism of fracture healing and may lead to the development of miRNA-based tissue engineering strategies to promote fracture healing.
Collapse
Affiliation(s)
- Takahiro Waki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Sang Yang Lee
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Takashi Iwakura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Yoshihiro Dogaki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Etsuko Okumachi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Keisuke Oe
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Masahiro Kurosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
195
|
Sato N, Ichikawa J, Wako M, Ohba T, Saito M, Sato H, Koyama K, Hagino T, Schoenecker JG, Ando T, Haro H. Thrombin induced by the extrinsic pathway and PAR-1 regulated inflammation at the site of fracture repair. Bone 2016; 83:23-34. [PMID: 26475502 DOI: 10.1016/j.bone.2015.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/30/2015] [Accepted: 10/11/2015] [Indexed: 02/07/2023]
Abstract
Thrombin (coagulation factor IIa) is a serine protease encoded by the F2 gene. Pro-thrombin (coagulation factor II) is cut to generate thrombin in the coagulation cascade that results in a reduction of blood loss. Procoagulant states that lead to activation of thrombin are common in bone fracture sites. However, its physiological roles and relationship with osteoblasts in bone fractures are largely unknown. We herein report various effects of thrombin on mouse osteoblastic MC3T3-E1 cells. MC3T3-E1 cells expressed proteinase-activated receptor 1 (PAR1), also known as the coagulation factor II receptor. They also produced monocyte chemoattractant protein (MCP-1), tissue factor (TF), MCSF and IL-6 upon thrombin stimulation through the PI3K-Akt and MEK-Erk1/2 pathways. Furthermore, MCP-1 obtained from thrombin-stimulated MC3T3-E1 cells induced migration by macrophage RAW264 cells. All these effects of thrombin on MC3T3-E1 cells were abolished by the selective non-peptide thrombin receptor inhibitor SCH79797. We also found that thrombin, PAR-1, MCP-1, TF as well as phosphorylated AKT and p42/44 were significantly expressed at the fracture site of mouse femoral bone. Collectively, thrombin/PAR-1 interaction regulated MCP-1, TF, MCSF and IL-6 production by MC3T3-E1 cells. Furthermore, MCP-1 induced RAW264 cell migration. Thrombin may thus be a novel cytokine that regulates several aspects of osteoblast function and fracture healing.
Collapse
Affiliation(s)
- Nobutaka Sato
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokatou, Chuo, Yamanashi 409-3898, Japan
| | - Jiro Ichikawa
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokatou, Chuo, Yamanashi 409-3898, Japan
| | - Masanori Wako
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokatou, Chuo, Yamanashi 409-3898, Japan
| | - Tetsuro Ohba
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokatou, Chuo, Yamanashi 409-3898, Japan
| | - Masanori Saito
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokatou, Chuo, Yamanashi 409-3898, Japan
| | - Hironao Sato
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokatou, Chuo, Yamanashi 409-3898, Japan
| | - Kensuke Koyama
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokatou, Chuo, Yamanashi 409-3898, Japan
| | - Tetsuo Hagino
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokatou, Chuo, Yamanashi 409-3898, Japan; The Sports Medicine and Knee Center, Kofu National Hospital, 11-35 Tenjincho, Kofu, Yamanashi 400-8533, Japan
| | - Jonathan G Schoenecker
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 2200 Children's Way, Nashville, TN 37232-9565, United States; Department of Orthopaedics, Vanderbilt University Medical Center, 2200 Children's Way, Nashville, TN 37232-9565, United States; Department of Center for Bone Biology, Vanderbilt University Medical Center, 2200 Children's Way, Nashville, TN 37232-9565, United States; Department of Pharmacology, Vanderbilt University Medical Center, 2200 Children's Way, Nashville, TN 37232-9565, United States; Department of Pediatrics, Vanderbilt University Medical Center, 2200 Children's Way, Nashville, TN 37232-9565, United States
| | - Takashi Ando
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokatou, Chuo, Yamanashi 409-3898, Japan.
| | - Hirotaka Haro
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, 1110 Shimokatou, Chuo, Yamanashi 409-3898, Japan
| |
Collapse
|
196
|
Chan JK, Glass GE, Ersek A, Freidin A, Williams GA, Gowers K, Espirito Santo AI, Jeffery R, Otto WR, Poulsom R, Feldmann M, Rankin SM, Horwood NJ, Nanchahal J. Low-dose TNF augments fracture healing in normal and osteoporotic bone by up-regulating the innate immune response. EMBO Mol Med 2016; 7:547-61. [PMID: 25770819 PMCID: PMC4492816 DOI: 10.15252/emmm.201404487] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mechanism by which trauma initiates healing remains unclear. Precise understanding of these events may define interventions for accelerating healing that could be translated to the clinical arena. We previously reported that addition of low-dose recombinant human TNF (rhTNF) at the fracture site augmented fracture repair in a murine tibial fracture model. Here, we show that local rhTNF treatment is only effective when administered within 24 h of injury, when neutrophils are the major inflammatory cell infiltrate. Systemic administration of anti-TNF impaired fracture healing. Addition of rhTNF enhanced neutrophil recruitment and promoted recruitment of monocytes through CCL2 production. Conversely, depletion of neutrophils or inhibition of the chemokine receptor CCR2 resulted in significantly impaired fracture healing. Fragility, or osteoporotic, fractures represent a major medical problem as they are associated with permanent disability and premature death. Using a murine model of fragility fractures, we found that local rhTNF treatment improved fracture healing during the early phase of repair. If translated clinically, this promotion of fracture healing would reduce the morbidity and mortality associated with delayed patient mobilization.
Collapse
Affiliation(s)
- James K Chan
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Graeme E Glass
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Adel Ersek
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew Freidin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Garry A Williams
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Kate Gowers
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Rosemary Jeffery
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - William R Otto
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - Richard Poulsom
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - Marc Feldmann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Sara M Rankin
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Nicole J Horwood
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | |
Collapse
|
197
|
Effect of Short-Term Stimulation with Interleukin-1β and Differentiation Medium on Human Mesenchymal Stromal Cell Paracrine Activity in Coculture with Osteoblasts. BIOMED RESEARCH INTERNATIONAL 2015; 2015:714230. [PMID: 26798640 PMCID: PMC4700155 DOI: 10.1155/2015/714230] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/10/2015] [Accepted: 11/26/2015] [Indexed: 01/28/2023]
Abstract
Introduction. Human mesenchymal stromal cells (hMSCs) exhibit the potential to accelerate bone healing by enhanced osteogenic differentiation. Interleukin-1β is highly expressed during fracture healing and has been demonstrated to exert a significant impact on the differentiation behaviour of hMSCs. Here, we investigate the effect of 2-hour IL-1β stimulation on the differentiation and paracrine activity of hMSCs in coculture with osteosarcoma cells in vitro. Methods. hMSCs from 3 donors were incubated for 2 hours with 10 ng/mL IL-1β and subsequently cocultured with MG63-GFP cells either in control or in differentiation medium in a transwell system for 28 days. Genetic and functional effects were investigated. Results. hMSCs cultured in control medium exhibited a regulatory effect on cocultured MG63-GFP cells, resulting in upregulation of osteogenic gene expression in combination with increased ALP activity. However, while stimulated hMSCs cultured under differentiation conditions exhibit signs of osteogenic differentiation, osteogenic differentiation also caused an impaired regulatory effect on the cocultured MG63-GFP cells. Conclusion. Short stimulation of hMSCs has the potential to modify their long-term behaviour. In addition, undifferentiated hMSCs are able to regulate osteoblast differentiation; however, this regulatory function is lost upon osteogenic differentiation in vitro. This offers a novel approach for clinical cell therapy protocols.
Collapse
|
198
|
Li Z, Kuhn G, von Salis-Soglio M, Cooke SJ, Schirmer M, Müller R, Ruffoni D. In vivo monitoring of bone architecture and remodeling after implant insertion: The different responses of cortical and trabecular bone. Bone 2015; 81:468-477. [PMID: 26303288 DOI: 10.1016/j.bone.2015.08.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 08/17/2015] [Accepted: 08/19/2015] [Indexed: 12/01/2022]
Abstract
The mechanical integrity of the bone-implant system is maintained by the process of bone remodeling. Specifically, the interplay between bone resorption and bone formation is of paramount importance to fully understand the net changes in bone structure occurring in the peri-implant bone, which are eventually responsible for the mechanical stability of the bone-implant system. Using time-lapsed in vivo micro-computed tomography combined with new composite material implants, we were able to characterize the spatio-temporal changes of bone architecture and bone remodeling following implantation in living mice. After insertion, implant stability was attained by a quick and substantial thickening of the cortical shell which counteracted the observed loss of trabecular bone, probably due to the disruption of the trabecular network. Within the trabecular compartment, the rate of bone formation close to the implant was transiently higher than far from the implant mainly due to an increased mineral apposition rate which indicated a higher osteoblastic activity. Conversely, in cortical bone, the higher rate of bone formation close to the implant compared to far away was mostly related to the recruitment of new osteoblasts as indicated by a prevailing mineralizing surface. The behavior of bone resorption also showed dissimilarities between trabecular and cortical bone. In the former, the rate of bone resorption was higher in the peri-implant region and remained elevated during the entire monitoring period. In the latter, bone resorption rate had a bigger value away from the implant and decreased with time. Our approach may help to tune the development of smart implants that can attain a better long-term stability by a local and targeted manipulation of the remodeling process within the cortical and the trabecular compartments and, particularly, in bone of poor health.
Collapse
Affiliation(s)
- Zihui Li
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Gisela Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | | | | | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Davide Ruffoni
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland; Department of Aerospace and Mechanical Engineering, University of Liege, Liege, Belgium.
| |
Collapse
|
199
|
Dhaliwal K, Kunchur R, Farhadieh R. Review of the cellular and biological principles of distraction osteogenesis: An in vivo bioreactor tissue engineering model. J Plast Reconstr Aesthet Surg 2015; 69:e19-26. [PMID: 26725979 DOI: 10.1016/j.bjps.2015.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 09/22/2015] [Accepted: 11/14/2015] [Indexed: 02/06/2023]
Abstract
Distraction osteogenesis (DO) is a widely used technique in plastic and orthopaedic surgery. During the process, mechanical force is applied to fractured bone to enhance the regenerative processes and induce new bone formation. Although there is an abundance of literature on the clinical process of DO, there is a distinct lack of focus on the underlying biological principles governing this process. DO follows the basic premises of tissue engineering. The mechanical stress stimulates mesenchymal stem cell differentiation down an osteoblastic lineage on a matrix background. The aim of this review is to give an overview of the current knowledge of the molecular mechanism governing this process.
Collapse
Affiliation(s)
- K Dhaliwal
- St George's NHS Trust, Tooting, London, SW17 0QT, UK.
| | - R Kunchur
- Plastic & Reconstructive Surgery Department, Australian National University, Canberra ACT 0200, Australia
| | - R Farhadieh
- Plastic & Reconstructive Surgery Department, Australian National University, Canberra ACT 0200, Australia
| |
Collapse
|
200
|
Schira J, Schulte M, Döbele C, Wallner C, Abraham S, Daigeler A, Kneser U, Lehnhardt M, Behr B. Human scaphoid non-unions exhibit increased osteoclast activity compared to adjacent cancellous bone. J Cell Mol Med 2015; 19:2842-50. [PMID: 26416438 PMCID: PMC4687713 DOI: 10.1111/jcmm.12677] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 08/03/2015] [Indexed: 01/08/2023] Open
Abstract
Scaphoid bones have a high prevalence for non-union. Even with adequate treatment, bone regeneration may not occur in certain instances. Although this condition is well described, the molecular pathology of scaphoid non-unions is still poorly defined. In this study, gene expression of osteogenic and angiogenic growth and transcription factors as well as inflammatory mediators were analysed in human scaphoid non-unions and intraindividually compared to adjacent autologous cancellous bone from the distal radius. In addition, histology and immunohistochemical stainings were performed to verify qRT-PCR data. Gene expression analysis revealed a significant up-regulation of RANKL, ALP, CYCLIN D1, MMP-13, OPG, NFATc1, TGF-β and WNT5A in scaphoid non-unions. Interestingly, RANKL and NFATc1, both markers for osteoclastogenesis, were significantly induced in non-unions. Moreover, WNT5A was highly up-regulated in all non-union samples. TRAP staining confirmed the observation of induced osteoclastogenesis in non-unions. With respect to genes related to osteogenesis, alkaline phosphatase was significantly up-regulated in scaphoid non-unions. No differences were detectable for other osteogenic genes such as RUNX-2 or BMP-2. Importantly, we did not detect differences in angiogenesis between scaphoid non-unions and controls in both gene expression and immunohistochemistry. Summarized, our data indicate increased osteoclast activity in scaphoid non-unions possibly as a result of the alterations in RANKL, TGF-β and WNT5A expression levels. These data increase our understanding for the reduced bone regeneration capacity present in scaphoid non-unions and may translate into the identification of new therapeutic targets to avoid secondary damages and prevent occurrence of non-unions to scaphoid bones.
Collapse
Affiliation(s)
- Jessica Schira
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Matthias Schulte
- Department of Plastic Surgery, BG Trauma Hospital Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Carmen Döbele
- Department of Plastic Surgery, BG Trauma Hospital Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Christoph Wallner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Stephanie Abraham
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Adrien Daigeler
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Ulrich Kneser
- Department of Plastic Surgery, BG Trauma Hospital Ludwigshafen, University of Heidelberg, Ludwigshafen, 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
|