1
|
Woloszyk A, Aguilar L, Perez L, Salinas EL, Glatt V. Biomimetic hematoma delivers an ultra-low dose of rhBMP-2 to successfully regenerate large femoral bone defects in rats. BIOMATERIALS ADVANCES 2023; 148:213366. [PMID: 36905826 DOI: 10.1016/j.bioadv.2023.213366] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/10/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023]
Abstract
Successful repair of large bone defects remains a clinical challenge. Following fractures, a bridging hematoma immediately forms as a crucial step that initiates bone healing. In larger bone defects the micro-architecture and biological properties of this hematoma are compromised, and spontaneous union cannot occur. To address this need, we developed an ex vivo Biomimetic Hematoma that resembles naturally healing fracture hematoma, using whole blood and the natural coagulants calcium and thrombin, as an autologous delivery vehicle for a very reduced dose of rhBMP-2. When implanted into a rat femoral large defect model, complete and consistent bone regeneration with superior bone quality was achieved with 10-20× less rhBMP-2 compared to that required with the collagen sponges currently used. Moreover, calcium and rhBMP-2 demonstrated a synergistic effect enhancing osteogenic differentiation, and fully restored mechanical strength 8 weeks after surgery. Collectively, these findings suggest the Biomimetic Hematoma provides a natural reservoir for rhBMP-2, and that retention of the protein within the scaffold rather than its sustained release might be responsible for more robust and rapid bone healing. Clinically, this new implant, using FDA-approved components, would not only reduce the risk of adverse events associated with BMPs, but also decrease treatment costs and nonunion rates.
Collapse
Affiliation(s)
- Anna Woloszyk
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA
| | - Leonardo Aguilar
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA
| | - Louis Perez
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA
| | - Emily L Salinas
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA
| | - Vaida Glatt
- Department of Orthopaedics, University of Texas Health Science Center, San Antonio 78229, TX, USA; Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio 78229, TX, USA.
| |
Collapse
|
2
|
Fibrin glue does not promote migration and proliferation of bone marrow derived mesenchymal stem cells in collagenic membranes: an in vitro study. Sci Rep 2022; 12:20660. [PMID: 36450814 PMCID: PMC9712600 DOI: 10.1038/s41598-022-25203-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
During Autologous Matrix-Induced Chondrogenesis (AMIC), the membrane is often glued into the chondral defect. However, whether fibrin glue influences cells proliferation and migration remain unclear. This study evaluated the impact of fibrin glue addition to biologic membranes loaded with bone marrow-derived mesenchymal stem cells (B-MSCs). A porcine derived collagen membrane (Cartimaix, Matricel GmbH, Germany) was used. B-MSCs were harvested from three different unrelated donors. The membranes were embedded in mounting medium with DAPI (ABCAM, Cambridge, UK) and analysed at 1-, 2-, 3-, 4-, 6-, and at 8-week follow-up. The DAPI ties the DNA of the cell nucleus, emitting blue fluorescence. DAPI/nuclei signals were analysed with fluorescence microscopy at 100-fold magnification. The group without fibrin glue demonstrated greater migration of the B-MSCs within the membrane at week 4 (P < 0.001), 6 (P < 0.001), and 8 (P < 0.001). No difference was found at week 1, 2, and 3. The group without fibrin glue demonstrated greater proliferation of B-MSCs within the membrane. These differences were significant at week 1 (P = 0.02), 2 (P = 0.008), 3 (P = 0.0009), 4 (P < 0.0001), 6 (P < 0.0001), 8 (P < 0.0001). Concluding, in the present setting, the use of fibrin in a collagenic biomembrane impairs B-MSCs proliferation and migration in vitro.
Collapse
|
3
|
Fixation of the Membrane during Matrix-Induced Autologous Chondrocyte Implantation in the Knee: A Systematic Review. Life (Basel) 2022; 12:life12111718. [PMID: 36362873 PMCID: PMC9698345 DOI: 10.3390/life12111718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: It is unclear whether the type of membrane used for matrix-assisted autologous chondrocyte implantation (mACI) influences results. A systematic review was conducted to investigate the midterm results of the three most common types of membrane fixation for mACI. Methods: This systematic review was conducted according to the 2020 PRISMA checklist. PubMed, Google Scholar, Embase, and Scopus online databases were accessed in August 2022. All the prospective clinical trials reporting outcomes of mACI in the knee were considered. Studies that describe the modality of membrane fixation (glued, glued, and sutured, no fixation) used for mACI were eligible. Studies that conducted a minimum of 12 months of follow-up were considered. The outcomes of interest were the Tegner Activity Scale and International Knee Documentation Committee (IKDC) score. The rate of failure and revisions were also collected. Results: Data from 26 studies (1539 procedures; 554 of 1539 (36%) were women) were retrieved. The mean follow-up was 42.6 (12 to 84) months. No difference between the groups was found in terms of mean duration of symptoms, age, BMI, gender, and defect size (P > 0.1). No difference was found in terms of the Tegner score (P = 0.3). When no fixation was used, a statistically significant higher IKDC compared to the other groups (P = 0.02) was evidenced. No difference was found in the rate of failure (P = 0.1). The no-fixation group evidenced a statistically significant lower rate of revisions (P = 0.02). Conclusions: No membrane fixation for mACI in the knee scored better than the fastening techniques at the midterm follow-up.
Collapse
|
4
|
Migliorini F, Prinz J, Maffulli N, Eschweiler J, Weber C, Lecoutrier S, Hildebrand F, Greven J, Schenker H. Fibrin glue does not assist migration and proliferation of chondrocytes in collagenic membranes: an in vitro study. J Orthop Surg Res 2022; 17:311. [PMID: 35690862 PMCID: PMC9188690 DOI: 10.1186/s13018-022-03201-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/31/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Some authors secured the membrane during matrix-induced autologous chondrocyte implantation (mACI) with fibrin glue or did not use a formal fixation. The real impact of fibrin glue addition on chondrocytes migration and proliferation has not yet been clarified. This study evaluated the impact of fibrin glue on a chondrocyte loaded collagenic membrane. METHODS A resorbable collagen I/III porcine derived membrane commonly employed in AMIC was used for all experiments. Chondrocytes from three difference donors were used. At 1-, 2-, 3-, 4-, 6-, and at 8-week the membranes were embedded in Mounting Medium with Dapi (ABCAM, Cambridge, UK). The Dapi contained in the mounting medium ties the DNA of the cell nucleus and emits a blue fluorescence. In this way, the spreading of the cells in the membrane can be easily monitored. The outcomes of interest were to evaluate (1) cell migration and (2) cell proliferation within the porous membrane layer. DAPI/nuclei signals were analysed with fluorescence microscope under a magnification of 100-fold. RESULTS The no-fibrin group demonstrated greater migration of the cells within the membrane. Although migration resulted higher in the no-fibrin group at every follow-up, this difference was significant only at week 1 (P < 0.001), 2 (P = 0.004), and 3 (P = 0.03). No difference was found at week 3, 6, and 8. The no-fibrin group demonstrated greater proliferation of the chondrocytes within the membrane. These differences were significant at week 4 (P < 0.0001), 6 (P < 0.0001), 8 (P < 0.0001). CONCLUSION The use of fibrin glue over a resorbable membrane leads to lower in vitro proliferation and migration of chondrocytes.
Collapse
Affiliation(s)
- Filippo Migliorini
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany.
| | - Julia Prinz
- Department of Ophthalmology, RWTH University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081, Baronissi, SA, Italy.
- School of Pharmacy and Bioengineering, Faculty of Medicine, Keele University, ST4 7QB, Stoke on Trent, England.
- Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, Mile End Hospital, 275 Bancroft Road, E1 4DG, London, England.
| | - Jörg Eschweiler
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Christian Weber
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Sophie Lecoutrier
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Frank Hildebrand
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Johannes Greven
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Hanno Schenker
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| |
Collapse
|
5
|
Migliorini F, Maffulli N, Baroncini A, Bell A, Hildebrand F, Schenker H. Autologous matrix-induced chondrogenesis is effective for focal chondral defects of the knee. Sci Rep 2022; 12:9328. [PMID: 35661147 PMCID: PMC9167289 DOI: 10.1038/s41598-022-13591-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/17/2022] [Indexed: 12/05/2022] Open
Abstract
Focal chondral defects of the knee are common and their management is challenging. This study investigated the efficacy and safety of Autologous Matrix-Induced Chondrogenesis (AMIC) for focal chondral defects of the knee. A systematic review and meta-analysis was conducted (according to the 2020 PRISMA statement) to investigate the efficacy of AMIC in improving symptoms and to compare AMIC versus microfracture (MFx). In January 2022, the following databases were accessed: Pubmed, Web of Science, Google Scholar, Embase. No time constrain was used for the search. All the clinical trials investigating AMIC and/or those comparing AMIC versus MFx for focal chondral defects of the knee were accessed. Only studies published in peer reviewed journals were considered. Studies which investigated other locations of the defects rather than knee were not eligible, nor those reporting data form mixed locations. Studies which reported data on revision settings, as well as those investigating efficacy on kissing lesions or multiple locations, were not suitable. The mean difference (MD) and odd ratio (OR) effect measure were used for continuous and binary data, respectively. Data from 18 studies (548 patients) were retrieved with a mean follow-up of 39.9 ± 26.5 months. The mean defect size was 3.2 ± 1.0 cm2. The visual analogue scale (VAS) decreased of − 3.9/10 (95% confidence interval (CI) − 4.0874 to -3.7126), the Tegner Activity Scale increased of + 0.8/10 (95% CI 0.6595 to 0.9405). The Lysholm Knee Scoring System increased of + 28.9/100 (95% CI 26.8716 to 29.1284), as did the International Knee Documentation Committee (IKDC) + 33.6/100 (95% CI 32.5800 to 34.6200). At last follow-up no patient showed signs of hypertrophy. 4.3% (9 of 210) of patients underwent revision procedures. The rate of failure was 3.8% (9 of 236). Compared to MFx, AMIC demonstrated lower VAS score (MD: − 1.01; 95% CI − 1.97 to 0.05), greater IKDC (MD: 11.80; 95% CI 6.65 to 16.94), and lower rate of revision (OR: 0.16; 95% CI 0.06 to 0.44). AMIC is effective for focal chondral defects of the knee. Furthermore, AMIC evidenced greater IKDC, along with a lower value of VAS and rate of revision compared to MFx.
Collapse
Affiliation(s)
- Filippo Migliorini
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany.
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081, Baronissi, SA, Italy.,School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, ST4 7QB, Stoke on Trent, England.,Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Mile End Hospital, E1 4DG, London, England
| | - Alice Baroncini
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Andreas Bell
- Department of Orthopaedic and Trauma Surgery, Eifelklinik St. Brigida, 52152, Simmerath, Germany
| | - Frank Hildebrand
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Hanno Schenker
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| |
Collapse
|
6
|
Migliorini F, Eschweiler J, Goetze C, Tingart M, Maffulli N. Membrane scaffolds for matrix-induced autologous chondrocyte implantation in the knee: a systematic review. Br Med Bull 2021; 140:50-61. [PMID: 34553227 DOI: 10.1093/bmb/ldab024] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 08/30/2021] [Accepted: 09/05/2021] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Chondral defects of the knee are common and their management is challenging. SOURCE OF DATA Current scientific literature published in PubMed, Google scholar, Embase and Scopus. AREAS OF AGREEMENT Membrane-induced autologous chondrocyte implantation (mACI) has been used to manage chondral defects of the knee. AREAS OF CONTROVERSY Hyaluronic acid membrane provides better outcomes than a collagenic membrane for mACI in the knee at midterm follow-up is controversial. GROWING POINTS To investigate whether hyaluronic acid membrane may provide comparable clinical outcomes than collagenic membranes for mACI in focal defects of the knee. AREAS TIMELY FOR DEVELOPING RESEARCH Hyaluronic acid membrane yields a lower rate of failures and revision surgeries for mACI in the management of focal articular cartilage defects of the knee compared with collagenic scaffolds at midterm follow-up. No difference was found in patient reported outcome measures (PROMs). Further comparative studies are required to validate these results in a clinical setting.
Collapse
Affiliation(s)
- Filippo Migliorini
- Department of Orthopaedic and Trauma Surgery, RWTH University Hospital Aachen, 52074 Aachen, Germany
| | - Jörg Eschweiler
- Department of Orthopaedic and Trauma Surgery, RWTH University Hospital Aachen, 52074 Aachen, Germany
| | - Christian Goetze
- Department of Orthopaedic Surgery, Auguste-Viktoria Clinic, Ruhr University Bochum, 32545, Bad Oeynhausen, Germany
| | - Markus Tingart
- Department of Orthopaedic and Trauma Surgery, RWTH University Hospital Aachen, 52074 Aachen, Germany
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, SA, Italy.,Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Mile End Hospital, 275 Bancroft Road, London E1 4DG, UK.,School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Thornburrow Drive, Stoke-on-Trent ST4 7QB, UK
| |
Collapse
|
7
|
Abstract
Hemophilia is caused by a lack of antihemophilic factor(s), for example, factor VIII (FVIII; hemophilia A) and factor IX (FIX; hemophilia B). Low bone mass is widely reported in epidemiological studies of hemophilia, and patients with hemophilia are at an increased risk of fracture. The detailed etiology of bone homeostasis imbalance in hemophilia is unclear. Clinical and experimental studies show that FVIII and FIX are involved in bone remodeling. However, it is likely that antihemophilic factors affect bone biology through thrombin pathways rather than via their own intrinsic properties. In addition, among patients with hemophilia, there are pathophysiological processes in several systems that might contribute to bone loss. This review summarizes studies on the association between hemophilia and bone remodeling, and might shed light on the challenges facing the care and prevention of osteoporosis and fracture in patients with hemophilia.
Collapse
Affiliation(s)
- Hanshi Wang
- Department of Sports Medicine and Joint Surgery, The People's Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xizhuang Bai
- Department of Sports Medicine and Joint Surgery, The People's Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
8
|
Huang H, Cheng WX, Hu YP, Chen JH, Zheng ZT, Zhang P. Relationship between heterotopic ossification and traumatic brain injury: Why severe traumatic brain injury increases the risk of heterotopic ossification. J Orthop Translat 2017; 12:16-25. [PMID: 29662775 PMCID: PMC5866497 DOI: 10.1016/j.jot.2017.10.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/12/2017] [Accepted: 10/18/2017] [Indexed: 01/01/2023] Open
Abstract
Heterotopic ossification (HO) is a pathological phenomenon in which ectopic lamellar bone forms in soft tissues. HO involves many predisposing factors, including congenital and postnatal factors. Postnatal HO is usually induced by fracture, burn, neurological damage (brain injury and spinal cord injury) and joint replacement. Recent studies have found that patients who suffered from bone fracture combined with severe traumatic brain injury (S-TBI) are at a significantly increased risk for HO occurrence. Thus, considerable research focused on the influence of S-TBI on fracture healing and bone formation, as well as on the changes in various osteogenic factors with S-TBI occurrence. Brain damage promotes bone formation, but the exact mechanisms underlying bone formation and HO after S-TBI remain to be clarified. Hence, this article summarises the findings of previous studies on the relationship between S-TBI and HO and discusses the probable causes and mechanisms of HO caused by S-TBI. The translational potential of this article: A better understanding of the probable causes of traumatic brain injury-induced HO can provide new perspectives and ideas in preventing HO and may support to design more targeted therapies to reduce HO or enhance the bone formation.
Collapse
Affiliation(s)
- Huan Huang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wen-Xiang Cheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi-Ping Hu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jian-Hai Chen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zheng-Tan Zheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Peng Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| |
Collapse
|
9
|
Kiernan J, Hu S, Grynpas MD, Davies JE, Stanford WL. Systemic Mesenchymal Stromal Cell Transplantation Prevents Functional Bone Loss in a Mouse Model of Age-Related Osteoporosis. Stem Cells Transl Med 2016; 5:683-93. [PMID: 26987353 DOI: 10.5966/sctm.2015-0231] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/23/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Age-related osteoporosis is driven by defects in the tissue-resident mesenchymal stromal cells (MSCs), a heterogeneous population of musculoskeletal progenitors that includes skeletal stem cells. MSC decline leads to reduced bone formation, causing loss of bone volume and the breakdown of bony microarchitecture crucial to trabecular strength. Furthermore, the low-turnover state precipitated by MSC loss leads to low-quality bone that is unable to perform remodeling-mediated maintenance--replacing old damaged bone with new healthy tissue. Using minimally expanded exogenous MSCs injected systemically into a mouse model of human age-related osteoporosis, we show long-term engraftment and markedly increased bone formation. This led to improved bone quality and turnover and, importantly, sustained microarchitectural competence. These data establish proof of concept that MSC transplantation may be used to prevent or treat human age-related osteoporosis. SIGNIFICANCE This study shows that a single dose of minimally expanded mesenchymal stromal cells (MSCs) injected systemically into a mouse model of human age-related osteoporosis display long-term engraftment and prevent the decline in bone formation, bone quality, and microarchitectural competence. This work adds to a growing body of evidence suggesting that the decline of MSCs associated with age-related osteoporosis is a major transformative event in the progression of the disease. Furthermore, it establishes proof of concept that MSC transplantation may be a viable therapeutic strategy to treat or prevent human age-related osteoporosis.
Collapse
Affiliation(s)
- Jeffrey Kiernan
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Sally Hu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Marc D Grynpas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - John E Davies
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - William L Stanford
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada Department of Cellular and Molecular Medicine and Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
10
|
Gur-Cohen S, Kollet O, Graf C, Esmon CT, Ruf W, Lapidot T. Regulation of long-term repopulating hematopoietic stem cells by EPCR/PAR1 signaling. Ann N Y Acad Sci 2016; 1370:65-81. [PMID: 26928241 DOI: 10.1111/nyas.13013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 01/05/2016] [Accepted: 01/11/2016] [Indexed: 01/18/2023]
Abstract
The common developmental origin of endothelial and hematopoietic cells is manifested by coexpression of several cell surface receptors. Adult murine bone marrow (BM) long-term repopulating hematopoietic stem cells (LT-HSCs), endowed with the highest repopulation and self-renewal potential, express endothelial protein C receptor (EPCR), which is used as a marker to isolate them. EPCR/protease-activated receptor-1 (PAR1) signaling in endothelial cells has anticoagulant and anti-inflammatory roles, while thrombin/PAR1 signaling induces coagulation and inflammation. Recent studies define two new PAR1-mediated signaling cascades that regulate EPCR(+) LT-HSC BM retention and egress. EPCR/PAR1 signaling facilitates LT-HSC BM repopulation, retention, survival, and chemotherapy resistance by restricting nitric oxide (NO) production, maintaining NO(low) LT-HSC BM retention with increased VLA4 expression, affinity, and adhesion. Conversely, acute stress and clinical mobilization upregulate thrombin generation and activate different PAR1 signaling that overcomes BM EPCR(+) LT-HSC retention, inducing their recruitment to the bloodstream. Thrombin/PAR1 signaling induces NO generation, TACE-mediated EPCR shedding, and upregulation of CXCR4 and PAR1, leading to CXCL12-mediated stem and progenitor cell mobilization. This review discusses new roles for factors traditionally viewed as coagulation related, which independently act in the BM to regulate PAR1 signaling in bone- and blood-forming progenitor cells, navigating their fate by controlling NO production.
Collapse
Affiliation(s)
- Shiri Gur-Cohen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Orit Kollet
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Claudine Graf
- Center for Thrombosis and Hemostasis and Johannes Gutenberg University Medical Center, Mainz, Germany.,Third Medical Department, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Charles T Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation and Departments of Pathology and Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis and Johannes Gutenberg University Medical Center, Mainz, Germany.,Department of Immunology and Microbial Science, the Scripps Research Institute, La Jolla, California
| | - Tsvee Lapidot
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
11
|
PAR1 signaling regulates the retention and recruitment of EPCR-expressing bone marrow hematopoietic stem cells. Nat Med 2015; 21:1307-17. [PMID: 26457757 DOI: 10.1038/nm.3960] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/31/2015] [Indexed: 12/14/2022]
Abstract
Retention of long-term repopulating hematopoietic stem cells (LT-HSCs) in the bone marrow is essential for hematopoiesis and for protection from myelotoxic injury. We report that signaling cascades that are traditionally viewed as coagulation related also control retention of endothelial protein C receptor-positive (EPCR(+)) LT-HSCs in the bone marrow and their recruitment to the blood via two pathways mediated by protease activated receptor 1 (PAR1). Thrombin-PAR1 signaling induces nitric oxide (NO) production, leading to EPCR shedding mediated by tumor necrosis factor-α-converting enzyme (TACE), enhanced CXCL12-CXCR4-induced motility and rapid stem and progenitor cell mobilization. Conversely, bone marrow blood vessels provide a microenvironment enriched with activated protein C (aPC) that retains EPCR(+) LT-HSCs by limiting NO generation, reducing Cdc42 activity and enhancing integrin VLA4 affinity and adhesion. Inhibition of NO production by aPC-EPCR-PAR1 signaling reduces progenitor cell egress from the bone marrow, increases retention of bone marrow NO(low) EPCR(+) LT-HSCs and protects mice from chemotherapy-induced hematological failure and death. Our study reveals new roles for PAR1 and EPCR in controlling NO production to balance maintenance and recruitment of bone marrow EPCR(+) LT-HSCs, with potential clinical relevance for stem cell transplantation.
Collapse
|
12
|
Beringer LT, Kiechel MA, Komiya Y, Donius AE, Habas R, Wegst UGK, Schauer CL. Osteoblast biocompatibility of novel chitosan crosslinker, hexamethylene-1,6-diaminocarboxysulfonate. J Biomed Mater Res A 2015; 103:3026-33. [DOI: 10.1002/jbm.a.35438] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 12/18/2014] [Accepted: 02/04/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Laura T. Beringer
- Department of Materials Science and Engineering; Drexel University; 3141 Chestnut Street Philadelphia Pennsylvania 19104
| | - Marjorie A. Kiechel
- Department of Materials Science and Engineering; Drexel University; 3141 Chestnut Street Philadelphia Pennsylvania 19104
| | - Yuko Komiya
- Department of Biology; Temple University; 1900 North 12th Street Philadelphia Pennsylvania 19122
| | - Amalie E. Donius
- Department of Materials Science and Engineering; Drexel University; 3141 Chestnut Street Philadelphia Pennsylvania 19104
| | - Raymond Habas
- Department of Biology; Temple University; 1900 North 12th Street Philadelphia Pennsylvania 19122
| | - Ulrike G. K. Wegst
- Thayer School of Engineering; Dartmouth College; 14 Engineering Drive Hanover New Hampshire 03755
| | - Caroline L. Schauer
- Department of Materials Science and Engineering; Drexel University; 3141 Chestnut Street Philadelphia Pennsylvania 19104
| |
Collapse
|
13
|
Chen TL, Lu HJ, Zhang XH, Wang SF, Bao HW, Tang DH. The activation and osteoinduction effects of thrombin in the maxillary sinus augmentation process: does thrombin only display activation effects on platelet-rich plasma? Int J Oral Maxillofac Surg 2015; 44:806-7. [PMID: 25670387 DOI: 10.1016/j.ijom.2015.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/08/2015] [Indexed: 11/29/2022]
Affiliation(s)
- T-L Chen
- Department of Periodontology, Oral Research Centre of CPLA, Hospital 411 of CPLA, Shanghai, China.
| | - H-J Lu
- Aerospace Engineering Medical College, Fourth Military Medical University, Xi'an, China
| | - X-H Zhang
- Oral Research Centre of CPLA, Hospital 411 of CPLA, Shanghai, China
| | - S-F Wang
- Naval Medical Research Institute, Shanghai, China
| | - H-W Bao
- Department of Orthopedics, Hospital 411 of CPLA, Shanghai, China
| | - D-H Tang
- Department of Clinical Experiment, Hospital 411 of CPLA, Shanghai, China
| |
Collapse
|
14
|
Srivastava S, Sharma K, Kumar N, Roy P. Bradykinin regulates osteoblast differentiation by Akt/ERK/NFκB signaling axis. J Cell Physiol 2014; 229:2088-105. [PMID: 24825463 DOI: 10.1002/jcp.24668] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 05/09/2014] [Indexed: 12/11/2022]
Abstract
Bradykinin (BK), a well known mediator of pain and inflammation, is also known to be involved in the process of bone resorption. The present study therefore evaluated the role of BK in osteoblast lineage commitment. Our data showed that BK inhibits the migration of bone marrow mesenchymal stem cells, but does not affect their viability. Moreover, BK also inhibits osteoblastic differentiation by significantly downregulating the levels of mRNAs for osteopontin, runX2, col24, osterix, osteocalcin genes and bone mineralization (P < 0.05). Further, BK was found to elicit the BK receptors (BDKR1 and BDKR2) mediated activation of ERK1/2 and Akt pathways, which finally led to the activation of NFκB. BK also promoted the osteoclast differentiation of bone marrow derived preosteoclast cells by upregulating the expression of c-fos, NFATC1, TRAP, clcn7, cathK, and OSCAR genes and increasing TRAP activity through NFκB pathway. In conclusion, our data suggest that BK decreases the differentiation of osteoblasts with concomitant increase in osteoclast formation and thus provides new insight into the mechanism of action of BK in modulating bone resorption.
Collapse
Affiliation(s)
- Swati Srivastava
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | | | | | | |
Collapse
|
15
|
Marques LF, Stessuk T, Camargo ICC, Sabeh Junior N, Santos LD, Ribeiro-Paes JT. Platelet-rich plasma (PRP): Methodological aspects and clinical applications. Platelets 2014; 26:101-13. [DOI: 10.3109/09537104.2014.881991] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
16
|
Srivastava S, Kumar N, Roy P. Role of ERK/NFκB in vanadium (IV) oxide mediated osteoblast differentiation in C3H10t1/2 cells. Biochimie 2014; 101:132-44. [PMID: 24440756 DOI: 10.1016/j.biochi.2014.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 01/06/2014] [Indexed: 10/25/2022]
Abstract
Vanadium (V) compounds are reported to have insulin mimicking action, which render them to show excellent osteogenic activity. In the current study we investigated the effect of various vanadium compounds on osteoblast differentiation of mouse mesenchymal stem cells, C3H10t1/2 cells, and analyzed the underlying mechanism of vanadium for this action. Our data showed that treatment of C3H10t1/2 cells with V (IV) oxide complex (at 7-25 μM concentrations) induced osteoblast differentiation maximally as compared to V2O5. On the other hand, ammonium vanadate was found to dampen the osteoblast differentiation process. Based on this data, V (IV) oxide was investigated further to analyze its probable mode of action as an osteoblastic agent. The key factors implicated in osteoblast differentiation i.e., NFκB, ERK ½, AP1 and CRE were examined in response to V (IV) oxide exposure. Exposure to V (IV) oxide caused 2- and 5-folds induction of luciferase activities in cells transfected with SRE-luc and NFκB-luc reporter vectors respectively (p < 0.05). Further, exposure to V (IV) oxide enhanced the phosphorylation of ERK ½, IκB and NFκBp65 proteins. In addition, RT-PCR analysis, alizarin red staining and immunoblot analysis showed that inhibition of osteoblast differentiation in presence of PD98059 and parthenolide (inhibitors of ERK and NFκB pathways respectively) was rescued in presence of V (IV) oxide. These results suggest that V (IV) oxide up regulates osteoblast differentiation through ERK and NFκB pathways and hence could be utilized as an agent for bone formation after further analysis and validation.
Collapse
Affiliation(s)
- Swati Srivastava
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | - Narender Kumar
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India.
| |
Collapse
|
17
|
Research progress in the mechanism of effect of PRP in bone deficiency healing. ScientificWorldJournal 2013; 2013:134582. [PMID: 23710132 PMCID: PMC3654280 DOI: 10.1155/2013/134582] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 02/27/2013] [Indexed: 12/19/2022] Open
Abstract
Platelet-rich plasma (PRP) therapy is a recently developed technique that uses a concentrated portion of autologous blood to try to improve and accelerate the healing of various tissues. There is a considerable interest in using these PRP products for the treatment used in bone deficiency healing. Because PRP products are safe and easy to prepare and administer, there has been increased attention toward using PRP in numerous clinical settings. The benefits of PRP therapy appear to be promising, and many investigators are exploring the ways in which this therapy can be used in the clinical setting. At present, the molecular mechanisms of bone defect repair studies have focused on three aspects of the inflammatory cytokines, growth factors and angiogenic factors. The role of PRP works mainly through these three aspects of bone repair. The purpose of this paper is to review the current evidence on the mechanism of the effect of PRP in bone deficiency healing.
Collapse
|
18
|
Su N, Chen M, Chen S, Li C, Xie Y, Zhu Y, Zhang Y, Zhao L, He Q, Du X, Chen D, Chen L. Overexpression of H1 calponin in osteoblast lineage cells leads to a decrease in bone mass by disrupting osteoblast function and promoting osteoclast formation. J Bone Miner Res 2013; 28:660-71. [PMID: 23044709 PMCID: PMC3716280 DOI: 10.1002/jbmr.1778] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 08/24/2012] [Accepted: 09/07/2012] [Indexed: 02/03/2023]
Abstract
H1 calponin (CNN1) is known as a smooth muscle-specific, actin-binding protein which regulates smooth muscle contractive activity. Although previous studies have shown that CNN1 has effect on bone, the mechanism is not well defined. To investigate the role of CNN1 in maintaining bone homeostasis, we generated transgenic mice overexpressing Cnn1 under the control of the osteoblast-specific 3.6-kb Col1a1 promoter. Col1a1-Cnn1 transgenic mice showed delayed bone formation at embryonic stage and decreased bone mass at adult stage. Morphology analyses showed reduced trabecular number, thickness and defects in bone formation. The proliferation and migration of osteoblasts were decreased in Col1a1-Cnn1 mice due to alterations in cytoskeleton. The early osteoblast differentiation of Col1a1-Cnn1 mice was increased, but the late stage differentiation and mineralization of osteoblasts derived from Col1a1-Cnn1 mice were significantly decreased. In addition to impaired bone formation, the decreased bone mass was also associated with enhanced osteoclastogenesis. Tartrate-resistant acid phosphatase (TRAP) staining revealed increased osteoclast numbers in tibias of 2-month-old Col1a1-Cnn1 mice, and increased numbers of osteoclasts co-cultured with Col1a1-Cnn1 osteoblasts. The ratio of RANKL to OPG was significantly increased in Col1a1-Cnn1 osteoblasts. These findings reveal a novel function of CNN1 in maintaining bone homeostasis by coupling bone formation to bone resorption.
Collapse
Affiliation(s)
- Nan Su
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Maomao Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Siyu Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Can Li
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ying Zhu
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yaozong Zhang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ling Zhao
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Qifen He
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiaolan Du
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
19
|
Xu C, Poh YKC, Roes I, O'Cearbhaill ED, Matthiesen ME, Mu L, Yang SY, Miranda-Nieves D, Irimia D, Karp JM. A portable chemotaxis platform for short and long term analysis. PLoS One 2012; 7:e44995. [PMID: 23028729 PMCID: PMC3444504 DOI: 10.1371/journal.pone.0044995] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 08/15/2012] [Indexed: 11/18/2022] Open
Abstract
Flow-based microfluidic systems have been widely utilized for cell migration studies given their ability to generate versatile and precisely defined chemical gradients and to permit direct visualization of migrating cells. Nonetheless, the general need for bulky peripherals such as mechanical pumps and tubing and the complicated setup procedures significantly limit the widespread use of these microfluidic systems for cell migration studies. Here we present a simple method to power microfluidic devices for chemotaxis assays using the commercially available ALZET® osmotic pumps. Specifically, we developed a standalone chemotaxis platform that has the same footprint as a multiwell plate and can generate well-defined, stable chemical gradients continuously for up to 7 days. Using this platform, we validated the short-term (24 hours) and long-term (72 hours) concentration dependent PDGF-BB chemotaxis response of human bone marrow derived mesenchymal stem cells.
Collapse
Affiliation(s)
- Chenjie Xu
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Cambridge, Massachusetts, United States of America
- Division of Bioengineering, School of Chemical & Biomedical Engineering, Nanyang Technological University, Singapore
| | - Yuk Kee C. Poh
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Cambridge, Massachusetts, United States of America
| | - Isaac Roes
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Cambridge, Massachusetts, United States of America
| | - Eoin D. O'Cearbhaill
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Cambridge, Massachusetts, United States of America
| | - Mads Emil Matthiesen
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Cambridge, Massachusetts, United States of America
| | - Luye Mu
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Cambridge, Massachusetts, United States of America
| | - Seung Yun Yang
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Cambridge, Massachusetts, United States of America
| | - David Miranda-Nieves
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Cambridge, Massachusetts, United States of America
| | - Daniel Irimia
- BioMEMS Resource Center, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Shriners Hospital for Children, and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jeffrey M. Karp
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT, Division of Health Sciences and Technology, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
20
|
Marchand C, Chen G, Tran-Khanh N, Sun J, Chen H, Buschmann MD, Hoemann CD. Microdrilled Cartilage Defects Treated with Thrombin-Solidified Chitosan/Blood Implant Regenerate a More Hyaline, Stable, and Structurally Integrated Osteochondral Unit Compared to Drilled Controls. Tissue Eng Part A 2012; 18:508-19. [DOI: 10.1089/ten.tea.2011.0178] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Catherine Marchand
- Institute of Biomedical Engineering, École Polytechnique de Montréal, Montréal, Quebec, Canada
| | - Gaoping Chen
- Department of Chemical Engineering, École Polytechnique de Montréal, Quebec, Canada
| | - Nicolas Tran-Khanh
- Department of Chemical Engineering, École Polytechnique de Montréal, Quebec, Canada
- Groupe de Recherche en Sciences et Technologies Biomédicales, École Polytechnique de Montréal, Quebec, Canada
| | - Jun Sun
- BioSyntech Canada, Inc., Laval, Quebec, Canada
| | - Hongmei Chen
- Department of Chemical Engineering, École Polytechnique de Montréal, Quebec, Canada
| | - Michael D. Buschmann
- Institute of Biomedical Engineering, École Polytechnique de Montréal, Montréal, Quebec, Canada
- Department of Chemical Engineering, École Polytechnique de Montréal, Quebec, Canada
- Groupe de Recherche en Sciences et Technologies Biomédicales, École Polytechnique de Montréal, Quebec, Canada
| | - Caroline D. Hoemann
- Institute of Biomedical Engineering, École Polytechnique de Montréal, Montréal, Quebec, Canada
- Department of Chemical Engineering, École Polytechnique de Montréal, Quebec, Canada
- Groupe de Recherche en Sciences et Technologies Biomédicales, École Polytechnique de Montréal, Quebec, Canada
| |
Collapse
|
21
|
Palma-Nicolás JP, López E, López-Colomé AM. Thrombin stimulates RPE cell motility by PKC-zeta- and NF-kappaB-dependent gene expression of MCP-1 and CINC-1/GRO chemokines. J Cell Biochem 2010; 110:948-67. [PMID: 20564194 DOI: 10.1002/jcb.22608] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Retinal pigment epithelial cells (RPE) are the major cell type involved in the pathogenesis of proliferative vitreoretinopathy (PVR), which involves the epithelial-mesenchymal transition, proliferation, and directional migration of transformed RPE cells to the vitreous upon RPE exposure to serum components, thrombin among them. Although the aqueous humor and vitreous of PVR patients contain high levels of chemokines, their possible involvement in PVR development has not been explored. We here analyzed the effect of thrombin on chemokine gene expression and its correlation with RPE cell migration using rat RPE cells in culture as a model system. We demonstrated that thrombin induces RPE cell migration through the dose-dependent stimulation of MCP1 and GRO expression/release, and the autocrine activation of CXCR-2 and CCR-2 chemokine receptors. Whereas inhibition of CXCR2 by Sb-225002 and of CCR2 by Rs-504393 partially prevented hirudin-sensitive cell migration, the joint inhibition of these receptors abolished thrombin effect, suggesting the contribution of distinct but coincident mechanisms. Thrombin effects were not modified by Ro-32-0432 inhibition of conventional/novel PKC isoenzymes or by the MAPkinase pathway inhibitor U0126. MCP1 and GRO expression/secretion, and cell migration were completely prevented by the inhibitory PKC-zeta pseudosubstrate and by the nuclear factor-kappa B (NF-kappaB) inhibitor BAY11-7082, but not by wortmannin inhibition of PI3K. Results show that signaling pathways leading to RPE cell migration differ from the MEK-ERK-PI3K-mediated promotion RPE of cell proliferation, both of which concur at the activation of PKC-zeta.
Collapse
Affiliation(s)
- José Prisco Palma-Nicolás
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Coyoacan, México, DF
| | | | | |
Collapse
|
22
|
Kim ES, Kim JJ, Park EJ. Angiogenic factor-enriched platelet-rich plasma enhances in vivo bone formation around alloplastic graft material. J Adv Prosthodont 2010; 2:7-13. [PMID: 21165181 PMCID: PMC2984511 DOI: 10.4047/jap.2010.2.1.7] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 03/02/2010] [Accepted: 03/09/2010] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Although most researchers agree that platelet-rich plasma (PRP) is a good source of autogenous growth factors, its effect on bone regeneration is still controversial. The purpose of this study was to evaluate whether increasing angiogenic factors in the human PRP to enhance new bone formation through rapid angiogenesis. MATERIAL AND METHODS In vitro, the human platelets were activated with application of shear stress, 20 µg/ml collagen, 2 mM CaCl(2) and 10U thrombin/1 × 10(9) platelets. Level of vascular endothelial growth factor (VEGF) and platelet microparticle (PMP) in the activated platelets were checked. In the animal study, human angiogenic factors-enriched PRP was tested in 28 athymic rat's cranial critical bone defects with β-TCP. Angiogenesis and osteogenesis were evaluated by laser Doppler perfusion imaging, histology, dual energy X-ray densinometry, and micro-computed tomography. RESULTS In vitro, this human angiogenic factors-enriched PRP resulted in better cellular proliferation and osteogenic differentiation. In vivo, increasing angiogenic potential of the PRP showed significantly higher blood perfusion around the defect and enhanced new bone formation around acellular bone graft material. CONCLUSION Angiogenic factor-enriched PRP leads to faster and more extensive new bone formation in the critical size bone defect. The results implicate that rapid angiogenesis in the initial healing period by PRP could be supposed as a way to overcome short term effect of the rapid angiogenesis.
Collapse
Affiliation(s)
- Eun-Seok Kim
- Department of Oral & Maxillofacial Surgery, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Jae-Jin Kim
- Department of Oral & Maxillofacial Surgery, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Eun-Jin Park
- Division of Prosthodontics, School of Medicine, Ewha Womans University, Seoul, Korea
| |
Collapse
|
23
|
Kurata T, Hayashi T, Yoshikawa T, Okamoto T, Yoshida K, Iino T, Uchida A, Suzuki K. Activated protein C stimulates osteoblast proliferation via endothelial protein C receptor. Thromb Res 2009; 125:184-91. [PMID: 19804899 DOI: 10.1016/j.thromres.2009.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 08/24/2009] [Accepted: 09/08/2009] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Bone is continually remodeled by the action of osteoblasts, osteocytes, and osteoclasts. Resting osteoblasts are able to proliferate and differentiate into mature osteoblasts when physiologically required, as after tissue injury. Activated protein C (APC) is a serine protease that functions in anticoagulation, anti-inflammation, anti-apoptosis, cell proliferation, and wound repair. In this study, we examined the effect of APC on osteoblast proliferation and differentiation. MATERIALS AND METHODS We examined the presence of protein C in human fracture hematoma by immunohistochemical staining. We then evaluated the effect of APC, diisopropyl fluorophosphate-inactivated APC (DIP-APC) or protein C zymogen on normal human osteoblast (NHOst) proliferation using tetrazolium salt assay in the presence or absence of aprotinin, hirudin, protein C, antibody against protein C, endothelial protein C receptor (EPCR) or protease-activated receptor (PAR)-1. Finally, activation of p44/42 MAP kinase was evaluated by Western blot analysis. RESULTS Both APC and DIP-APC increased osteoblast proliferation in a dose-dependent manner, while protein C did not. The APC-induced increased proliferation of osteoblast was not affected by aprotinin, hirudin, and anti-protein C antibody which inhibits the protease activity of APC. Treatment with protein C or anti-EPCR antibody which inhibits APC binding to EPCR inhibited APC-mediated osteoblast proliferation, while treatment with anti-PAR-1 antibody did not. APC promoted the phosphorylation of p44/42 MAP kinase within osteoblasts; this effect was inhibited by the anti-EPCR antibody. CONCLUSIONS APC stimulates osteoblast proliferation by activating p44/42 MAP kinase through a mechanism that requires EPCR but not PAR-1 or the proteolytic activity of APC. APC generated at fracture sites may contribute to fracture healing by promoting osteoblast proliferation.
Collapse
Affiliation(s)
- Tatsuya Kurata
- Department of Molecular Pathobiology, Mie University Graduate School of Medicine, Tsu-city, Mie 514-8507, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Marchand C, Rivard GE, Sun J, Hoemann CD. Solidification mechanisms of chitosan-glycerol phosphate/blood implant for articular cartilage repair. Osteoarthritis Cartilage 2009; 17:953-60. [PMID: 19152788 DOI: 10.1016/j.joca.2008.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Revised: 10/24/2008] [Accepted: 12/06/2008] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Chitosan-glycerol phosphate (chitosan-GP) is a unique polymer solution that is mixed with whole blood and solidified over microfractured or drilled articular cartilage defects in order to elicit a more hyaline repair cartilage. For clinical ease-of-use, a faster in situ solidification is preferred. Therefore, we investigated the mechanisms underlying chitosan-GP/blood implant solidification. METHODS In vitro solidification of chitosan-GP/blood mixtures, with or without added clotting factors, was evaluated by thromboelastography. Serum was analyzed for the onset of thrombin, platelet, and FXIII activation. In vivo solidification of chitosan-GP/blood mixtures, with and without clotting factors, was evaluated in microdrilled cartilage defects of adult rabbits (N=41 defects). RESULTS Chitosan-GP/blood clots solidified in an atypical biphasic manner, with higher initial viscosity and minor platelet activation followed by the development of clot tensile strength concomitant with thrombin generation, burst platelet and FXIII activation. Whole blood and chitosan-GP/blood clots developed a similar final clot tensile strength, while polymer-blood clots showed a unique, sustained platelet factor release and greater resistance to lysis by tissue plasminogen activator. Thrombin, tissue factor (TF), and recombinant human activated factor VII (rhFVIIa) accelerated chitosan-GP/blood solidification in vitro (P<0.05). Pre-application of thrombin or rhFVIIa+TF to the surface of drilled cartilage defects accelerated implant solidification in vivo (P<0.05). CONCLUSIONS Chitosan-GP/blood implants solidify through coagulation mechanisms involving thrombin generation, platelet activation and fibrin polymerization, leading to a dual fibrin-polysaccharide clot scaffold that resists lysis and is physically more stable than normal blood clots. Clotting factors have the potential to enhance the practical use, the residency, and therapeutic activity of polymer-blood implants.
Collapse
Affiliation(s)
- C Marchand
- Institute of Biomedical Engineering, Ecole Polytechnique, Montreal, QC, Canada
| | | | | | | |
Collapse
|
25
|
Han B, Woodell-May J, Ponticiello M, Yang Z, Nimni M. The effect of thrombin activation of platelet-rich plasma on demineralized bone matrix osteoinductivity. J Bone Joint Surg Am 2009; 91:1459-70. [PMID: 19487525 DOI: 10.2106/jbjs.h.00246] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Demineralized bone matrix is an osteoinductive and osteoconductive material that is often used in orthopaedic surgery to induce bone formation. Autologous platelet-rich plasma, which contains proliferative and chemoattractant growth factors, has been used as a demineralized bone matrix adjuvant with mixed results. One variable during clinical use appears to be whether the platelet-rich plasma is activated with thrombin or is implanted in a liquid form with intact platelets. The objective of the present study was to determine if platelet-rich plasma can increase the osteoinductivity of demineralized bone matrix when used without thrombin activation. METHODS The bioactivity of the demineralized bone matrix was evaluated in vitro by determining alkaline phosphatase production by C2C12 myoblast cells. The effect of thrombin activation on platelet-rich plasma was studied in vitro by evaluating osteosarcoma and bone marrow stromal cells for cell number and transforming growth factor-beta1 activation. Demineralized bone matrices supplemented with platelet-rich plasma, with or without thrombin activation, were implanted intramuscularly in athymic rats and were examined at fourteen, twenty-eight, and fifty-six days. Histological samples were analyzed for osteogenesis and chondrogenesis. Osteogenesis was further characterized on the basis of alkaline phosphatase activity. RESULTS In vitro, thrombin triggered an immediate release of growth factors from the platelet-rich plasma, and the platelet-rich plasma increased the number of both osteosarcoma and stromal cells in a dose-dependent manner. Thrombin activation of the platelet-rich plasma eliminated such stimulatory effects. In vivo, the platelet-rich plasma stimulated chondrogenesis on Day 14 and osteogenesis on Days 28 and 56, whereas thrombin-activated platelet-rich plasma acted as an inhibitor of such events. In addition, inflammatory cells were detected in demineralized bone matrix samples that were mixed with thrombin-activated platelet-rich plasma. These cells were not present in matrix mixed with platelet-rich plasma alone. CONCLUSIONS Platelet-rich plasma significantly increased in vivo demineralized bone matrix osteoinductivity only when used without thrombin activation.
Collapse
Affiliation(s)
- Bo Han
- Department of Surgery, University of Southern California, 1840 North Soto Street EDM-191, Los Angeles, CA 90032, USA.
| | | | | | | | | |
Collapse
|
26
|
Wang JJ, Ye F, Cheng LJ, Shi YJ, Bao J, Sun HQ, Wang W, Zhang P, Bu H. Osteogenic differentiation of mesenchymal stem cells promoted by overexpression of connective tissue growth factor. J Zhejiang Univ Sci B 2009; 10:355-367. [PMID: 19434762 PMCID: PMC2676415 DOI: 10.1631/jzus.b0820252] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Accepted: 01/19/2009] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Large segmental bone defect repair remains a clinical and scientific challenge with increasing interest focusing on combining gene transfection with tissue engineering techniques. The aim of this study is to investigate the effect of connective tissue growth factor (CTGF) on the proliferation and osteogenic differentiation of the bone marrow mesenchymal stem cells (MSCs). METHODS A CTGF-expressing plasmid (pCTGF) was constructed and transfected into MSCs. Then expressions of bone morphogenesis-related genes, proliferation rate, alkaline phosphatase activity, and mineralization were examined to evaluate the osteogenic potential of the CTGF gene-modified MSCs. RESULTS Overexpression of CTGF was confirmed in pCTGF-MSCs. pCTGF transfection significantly enhanced the proliferation rates of pCTGF-MSCs (P<0.05). CTGF induced a 7.5-fold increase in cell migration over control (P<0.05). pCTGF transfection enhanced the expression of bone matrix proteins, such as bone sialoprotein, osteocalcin, and collagen type I in MSCs. The levels of alkaline phosphatase (ALP) activities of pCTGF-MSCs at the 1st and 2nd weeks were 4.0- and 3.0-fold higher than those of MSCs cultured in OS-medium, significantly higher than those of mock-MSCs and normal control MSCs (P<0.05). Overexpression of CTGF in MSCs enhanced the capability to form mineralized nodules. CONCLUSION Overexpression of CTGF could improve the osteogenic differentiation ability of MSCs, and the CTGF gene-modified MSCs are potential as novel cell resources of bone tissue engineering.
Collapse
Affiliation(s)
- Jin-jing Wang
- Key Laboratory of Transplant Engineering and Immunology of Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Ye
- Key Laboratory of Transplant Engineering and Immunology of Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li-jia Cheng
- Key Laboratory of Transplant Engineering and Immunology of Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu-jun Shi
- Key Laboratory of Transplant Engineering and Immunology of Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ji Bao
- Key Laboratory of Transplant Engineering and Immunology of Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huai-qiang Sun
- Key Laboratory of Transplant Engineering and Immunology of Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Wang
- Key Laboratory of Transplant Engineering and Immunology of Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peng Zhang
- Key Laboratory of Transplant Engineering and Immunology of Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
27
|
Arayatrakoollikit U, Pavasant P, Yongchaitrakul T. Thrombin induces osteoprotegerin synthesis via phosphatidylinositol 3'-kinase/mammalian target of rapamycin pathway in human periodontal ligament cells. J Periodontal Res 2008; 43:537-43. [PMID: 18565131 DOI: 10.1111/j.1600-0765.2007.01071.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Thrombin influences the biological behavior of periodontal ligament cells and plays multiple roles in the early stages of bone healing. Osteoprotegerin (OPG) is one of the key molecules that regulate bone homeostasis and prevent osteoclastogenesis. The purpose of this study was to evaluate the biological effects of thrombin on OPG synthesis in human periodontal ligament (HPDL) cells in vitro. MATERIAL AND METHODS Cells were treated with various concentrations (0.001, 0.01 and 0.1 U/mL) of thrombin. The mRNA expression and protein synthesis of OPG, as well as of receptor activator of nuclear factor kappaB ligand (RANKL), were determined by reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis, respectively. The influence of thrombin on OPG synthesis and its signaling pathway were investigated using inhibitors. RESULTS Thrombin profoundly induces protein synthesis of OPG at 0.1 U/mL. The inductive effect was inhibited by cycloheximide, but not by indomethacin. The phosphatidylinositol 3'-kinase (PI3K) inhibitor, LY294002, and the mammalian target of rapamycin (mTOR) inhibitor, rapamycin, exerted an inhibitory effect on the thrombin-induced OPG synthesis. In addition, the effect was inhibited by protease-activated receptor (PAR)-1 antagonist. Activation of phospho-Akt (p-Akt) was observed and the effect was abolished by LY294002. CONCLUSION Thrombin induces OPG synthesis in HPDL cells post-transcriptionally, possibly through PAR-1. The regulation was through the PI3K/Akt and mTOR pathway. This finding suggests that thrombin may play a significant role in alveolar bone repair and homeostasis of periodontal tissue, partly through the OPG/RANKL system.
Collapse
Affiliation(s)
- U Arayatrakoollikit
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | | | | |
Collapse
|
28
|
Thibault MM, Hoemann CD, Buschmann MD. Fibronectin, vitronectin, and collagen I induce chemotaxis and haptotaxis of human and rabbit mesenchymal stem cells in a standardized transmembrane assay. Stem Cells Dev 2007; 16:489-502. [PMID: 17610379 DOI: 10.1089/scd.2006.0100] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The mesenchymal stem cell (MSC) is a critical element in tissue repair and regeneration. Its ability to differentiate into multiple connective tissue cell types and to self-renew has made it a prime candidate in regenerative medicine strategies. Currently, the environmental cues responsible for in situ recruitment and control of MSC distribution at repair sites are not entirely revealed and in particular the role of extracellular matrix (ECM) proteins as motogenic factors has not been studied. Here we have used a standardized transmembrane chemotaxis assay to assess the chemotactic and haptotactic potential of fibronectin, vitronectin, and collagen type 1 on MSCs from both rabbit and human origin. The use of both cell types was based in part on the widespread use of rabbit models for musculoskeletal-related tissue engineering and repair models and their unknown correspondence to human in terms of MSC migration. The optimized assay yielded a greatly increased chemotactic response toward known factors such as platelet-derived growth factor-BB (PDGF)-BB compared to previous studies. Our primary finding was that all three ECM proteins tested (fibronectin, vitronectin, and collagen I) induced significant motogenic activity, in both soluble and insoluble forms, for both rabbit and human MSCs. These results suggest that ECM proteins could play roles as significant as cytokines in the recruitment of pluripotential repair cells wound and tissue repair sites. Furthermore, designed ECM coatings of scaffolds or implants could provide a new tool to control both cell influx and outflux from the scaffold post-implantation. Finally, the similarity of motogenic behavior of both rabbit and human cells suggests the rabbit is a reliable model for assessing MSC recruitment in repair and regeneration strategies.
Collapse
Affiliation(s)
- Marc M Thibault
- Department of Chemical Engineering, Institute of Biomedical Engineering, Ecole Polytechnique, Montreal, Quebec, Canada
| | | | | |
Collapse
|
29
|
Coleman LS. A hypothesis: factor VII governs clot formation, tissue repair and apoptosis. Med Hypotheses 2007; 69:903-7. [PMID: 17383108 DOI: 10.1016/j.mehy.2007.01.063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Accepted: 01/24/2007] [Indexed: 11/30/2022]
Abstract
A hypothesis: thrombin is a "Universal Enzyme of Energy Transduction" that employs ATP energy in flowing blood to activate biochemical reactions and cell effects in both hemostasis and tissue repair. All cells possess PAR-1 (thrombin) receptors and are affected by thrombin elevations, and thrombin effects on individual cell types are determined by their unique complement of PAR-1 receptors. Disruption of the vascular endothelium (VE) activates a tissue repair mechanism (TRM) consisting of the VE, tissue factor (TF), and circulating Factors VII, IX and X that governs localized thrombin elevations to activate clot formation and cellular effects that repair tissue damage. The culmination of the repair process occurs with the restoration of the VE followed by declines in thrombin production that causes Apoptosis ("programmed cell death") in wound-healing fibroblasts, which functions as a mechanism to draw wound edges together. The location and magnitude of TRM activity governs the location and magnitude of Factor VIII activity and clot formation, but the large size of Factor VIII prevents it from penetrating the clot formed by its activity, so that its effects are self-limiting. Factors VII, IX and X function primarily as tissue repair enzymes, while Factor VIII and Factor XIII are the only serine protease enzymes in the "Coagulation Cascade" that are exclusively associated with hemostasis.
Collapse
|
30
|
Pagel CN, Sivagurunathan S, Loh LH, Tudor EM, Pike RN, Mackie EJ. Functional responses of bone cells to thrombin. Biol Chem 2006; 387:1037-41. [PMID: 16895473 DOI: 10.1515/bc.2006.128] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractCells responsible for the formation and maintenance of bone express thrombin-responsive members of the protease-activated receptor family of G protein-coupled receptors. Thrombin has been shown to elicit a number of functional responses in these cells, including proliferation and cytokine production in osteoblasts. Many, but not all, of the effects of thrombin on bone cells are initiated by activation of protease-activated receptor-1. A combination ofin vitroobservations and results ofin vivostudies in protease-activated receptor-1-null mice suggest that thrombin plays multiple roles in the early stages of bone healing.
Collapse
Affiliation(s)
- Charles N Pagel
- School of Veterinary Science, University of Melbourne, Parkville, Victoria 3010, Australia.
| | | | | | | | | | | |
Collapse
|
31
|
Bluteau G, Pilet P, Bourges X, Bilban M, Spaethe R, Daculsi G, Guicheux J. The modulation of gene expression in osteoblasts by thrombin coated on biphasic calcium phosphate ceramic. Biomaterials 2006; 27:2934-43. [PMID: 16436294 DOI: 10.1016/j.biomaterials.2006.01.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2005] [Accepted: 01/03/2006] [Indexed: 10/25/2022]
Abstract
For many years, fibrin sealants were associated with bone substitutes to promote bone healing. However, the osteoblastic response to fibrin sealant components remains poorly documented. In this study, MC3T3-E1 osteoblastic cells were cultured on biphasic calcium phosphate ceramic (MBCP) coated with Tissucol components (thrombin and fibrinogen). Analysis of osteoblastic differentiation markers by RT-PCR revealed that MBCP coated with Tissucol stimulated mRNA levels for osteocalcin and alkaline phosphatase (ALP). Of all the components of Tissucol, thrombin has been reported to affect osteoblastic behavior. Our results demonstrated that low thrombin concentrations (0.5-5 U/ml) stimulated mRNA levels for ALP, whereas high thrombin concentrations (50-100 U/ml) decreased mRNA levels for ALP and PTH/PTHrP receptor and also increased mRNA level for the osteoclastogenesis inhibitor OPG. As thrombin stimulated angiogenesis, we then wondered whether thrombin could influence the expression of angiogenic factors. Low thrombin concentrations were shown to up-regulate mRNA levels for VEGF-B and VEGF-R1, suggesting an autocrine/paracrine role for VEGF-B. Higher thrombin concentrations also up-regulated mRNA for VEGF-A and neuropilin-1. In conclusion, the association of MBCP with thrombin and fibrinogen appears to be a convenient scaffold for bone cell differentiation. Thrombin could also acts at the cellular level by increasing the angiogenic potential of osteoblasts as well as their responsiveness to thrombin and VEGF.
Collapse
Affiliation(s)
- Gilles Bluteau
- INSERM UMRS 791, Laboratory of Osteoarticular and Dental Tissue Engineering, University of Nantes, School of Dental Surgery, 1 Place Alexis Ricordeau, 44042 Nantes cedex 1, France
| | | | | | | | | | | | | |
Collapse
|