1
|
Lee HY, An SB, Hwang SY, Hwang GY, Lee HL, Park HJ, Shin J, Kim KN, Wee SW, Yoon SL, Ha Y. Synergistic enhancement of spinal fusion in preclinical models using low-dose rhBMP-2 and stromal vascular fraction in an injectable hydrogel composite. Mater Today Bio 2025; 30:101379. [PMID: 39759847 PMCID: PMC11699625 DOI: 10.1016/j.mtbio.2024.101379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
Spinal fusion surgery remains a significant challenge due to limitations in current bone graft materials, particularly in terms of bioactivity, integration, and safety. This study presents an innovative approach using an injectable hydroxyapatite/β-tricalcium phosphate (HA/β-TCP) hydrogel combined with stromal vascular fraction (SVF) and low-dose recombinant human BMP-2 (rhBMP-2) to enhance osteodifferentiation and angiogenesis. Through a series of in vitro studies and preclinical models involving rats and minipigs, we demonstrated that the hydrogel system enables the sustained release of rhBMP-2, resulting in significantly improved bone density and integration, alongside reduced inflammatory responses. The combination of rhBMP-2 and SVF in this injectable formulation yielded superior spinal fusion outcomes, with enhanced mechanical properties and increased bone mass in both small and large animal models. These findings suggest that this strategy offers a promising and safer alternative for spinal fusion, with strong potential for clinical application.
Collapse
Affiliation(s)
- Hye Yeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seong Bae An
- Department of Neurosurgery, School of Medicine, CHA University, CHA Bundang Medical Center, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Graduate School, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sae Yeon Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Gwang Yong Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Jung Park
- Department of Research Center, CGBio., co. Ltd, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Joongkyum Shin
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Keung Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung Won Wee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sol Lip Yoon
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| |
Collapse
|
2
|
Xiao J, Cao Y, Li X, Xu L, Wang Z, Huang Z, Mu X, Qu Y, Xu Y. Elucidation of Factors Affecting the Age-Dependent Cancer Occurrence Rates. Int J Mol Sci 2024; 26:275. [PMID: 39796131 PMCID: PMC11720044 DOI: 10.3390/ijms26010275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Cancer occurrence rates exhibit diverse age-related patterns, and understanding them may shed new and important light on the drivers of cancer evolution. This study systematically analyzes the age-dependent occurrence rates of 23 carcinoma types, focusing on their age-dependent distribution patterns, the determinants of peak occurrence ages, and the significant difference between the two genders. According to the SEER reports, these cancer types have two types of age-dependent occurrence rate (ADOR) distributions, with most having a unimodal distribution and a few having a bimodal distribution. Our modeling analyses have revealed that (1) the first type can be naturally and simply explained using two age-dependent parameters: the total number of stem cell divisions in an organ from birth to the current age and the availability levels of bloodborne growth factors specifically needed by the cancer (sub)type, and (2) for the second type, the first peak is due to viral infection, while the second peak can be explained as in (1) for each cancer type. Further analyses indicate that (i) the iron level in an organ makes the difference between the male and female cancer occurrence rates, and (ii) the levels of sex hormones are the key determinants in the onset age of multiple cancer types. This analysis deepens our understanding of the dynamics of cancer evolution shared by diverse cancer types and provides new insights that are useful for cancer prevention and therapeutic strategies, thereby addressing critical gaps in the current paradigm of oncological research.
Collapse
Affiliation(s)
- Jun Xiao
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Yangkun Cao
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
- School of Artificial Intelligence, Jilin University, Changchun 130012, China
| | - Xuan Li
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Long Xu
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Zhihang Wang
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Zhenyu Huang
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Xuechen Mu
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
- School of Mathematics, Jilin University, Changchun 130012, China
| | - Yinwei Qu
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Ying Xu
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| |
Collapse
|
3
|
Karavasili C, Young T, Francis J, Blanco J, Mancini N, Chang C, Bernstock JD, Connolly ID, Shankar GM, Traverso G. Local drug delivery challenges and innovations in spinal neurosurgery. J Control Release 2024; 376:1225-1250. [PMID: 39505215 DOI: 10.1016/j.jconrel.2024.10.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
The development of novel therapeutics in the field of spinal neurosurgery faces a litany of translational challenges. Achieving precise drug targeting within the confined spaces associated with the spinal cord, canal and vertebra requires the development of next generation delivery systems and devices. These must be capable of overcoming inherent barriers related to drug diffusion, whilst concurrently ensuring optimal drug distribution and retention. In this review, we provide an overview of the most recent advances in the therapeutic management of diseases and disorders affecting the spine, including systems and devices capable of releasing small molecules and biopharmaceuticals that help eliminate pain and restore the mechanical function and stability of the spine. We highlight material-based approaches and minimally invasive techniques that can be employed to provide control over drug release kinetics and improve retention. We also seek to explore how the newest advancements in nanotechnology, biomaterials, additive manufacturing technologies and imaging modalities can be employed in this translational pursuit. Finally, we discuss the landscape of clinical trials and recently approved products aimed at overcoming the complexities associated with drug delivery to the spine.
Collapse
Affiliation(s)
- Christina Karavasili
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Thomas Young
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Joshua Francis
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Julianna Blanco
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Nicholas Mancini
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Charmaine Chang
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Joshua D Bernstock
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ian D Connolly
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ganesh M Shankar
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni Traverso
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
4
|
Munsch MA, Dalton J, Chen SR, Tang M, Como CJ, Whaley JD, Sadhwani SD, Fourman MS, Shaw JD, Lee JY. Off-Label Bone Morphogenetic Protein 2 Use Results in Successful Posterolateral Lumbar Fusion in a Veteran Population. J Am Acad Orthop Surg Glob Res Rev 2024; 8:01979360-202410000-00006. [PMID: 39392937 PMCID: PMC11469840 DOI: 10.5435/jaaosglobal-d-23-00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 06/19/2024] [Accepted: 07/27/2024] [Indexed: 10/13/2024]
Abstract
INTRODUCTION Patients within the US Veterans Health Administration (VA) system have higher rates of comorbidities and chronic pain, increasing risks of complications/poor outcomes following spine surgery. Although the use of bone morphogenetic protein 2 (BMP-2) is established for anterior lumbar interbody fusion, its indications for off-label use in posterolateral fusion are unclear. The objective of this study was to evaluate safety and utility of BMP-2 in posterolateral fusion through a 15-year experience at the VA. METHODS Patients underwent posterolateral lumbosacral fusions with BMP-2 by a single VA surgeon from January 1, 2005, to January 1, 2020. The primary outcome was fusion assessed through postoperative radiographs. Secondary outcomes included adjacent segment disease (ASD) and postoperative pain clinic utilization. RESULTS Sixty-eight patients underwent lumbosacral posterolateral fusion with BMP-2; 77.9% were discharged home and had no postoperative complications. All patients achieved bony fusion at a mean of 113.3 ± 59.9 days postoperatively. Five patients were diagnosed with cancer postoperatively, and eight patients required revision for ASD. No notable predictors of ASD exist. Preoperative opioid use predicted postoperative pain clinic utilization. DISCUSSION Posterolateral lumbar fusion with BMP-2 in veterans yields high fusion rates and favorable complication profiles and should be considered in multimorbid hosts. STUDY DESIGN Retrospective review of prospectively collected data.
Collapse
Affiliation(s)
- Maria A Munsch
- From the Department of Orthopaedic Surgery, Division of Spine Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA (Dr. Munsch, Dr. Dalton, Dr. Chen, Dr. Tang, Dr. Como, Dr. Whaley, Dr. Sadhwani, Dr. Fourman, Dr. Shaw, and Dr. Lee); Department of Surgery, Orthopaedic Surgery Service, VA Pittsburgh Medical Center, Pittsburgh, PA (Dr. Munsch, Dr. Dalton, Dr. Chen, Dr. Fourman, Dr. Shaw, and Dr. Lee); the Pittsburgh Orthopaedic Spine Research Group (POSR), Pittsburgh, PA (Dr. Munsch, Dr. Dalton, Dr. Chen, Dr. Tang, Dr. Como, Dr. Whaley, Dr. Shaw, and Dr. Lee); and the Orland Bethel Family Musculoskeletal Research Center (BMRC), Pittsburgh, PA (Dr. Chen, Dr. Tang, Dr. Como, Dr. Shaw, and Dr. Lee)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
D'Souza RS, Her YF, Hussain N, Karri J, Schatman ME, Calodney AK, Lam C, Buchheit T, Boettcher BJ, Chang Chien GC, Pritzlaff SG, Centeno C, Shapiro SA, Klasova J, Grider JS, Hubbard R, Ege E, Johnson S, Epstein MH, Kubrova E, Ramadan ME, Moreira AM, Vardhan S, Eshraghi Y, Javed S, Abdullah NM, Christo PJ, Diwan S, Hassett LC, Sayed D, Deer TR. Evidence-Based Clinical Practice Guidelines on Regenerative Medicine Treatment for Chronic Pain: A Consensus Report from a Multispecialty Working Group. J Pain Res 2024; 17:2951-3001. [PMID: 39282657 PMCID: PMC11402349 DOI: 10.2147/jpr.s480559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Injectable biologics have not only been described and developed to treat dermal wounds, cardiovascular disease, and cancer, but have also been reported to treat chronic pain conditions. Despite emerging evidence supporting regenerative medicine therapy for pain, many aspects remain controversial. Methods The American Society of Pain and Neuroscience (ASPN) identified the educational need for an evidence-based guideline on regenerative medicine therapy for chronic pain. The executive board nominated experts spanning multiple specialties including anesthesiology, physical medicine and rehabilitation, and sports medicine based on expertise, publications, research, and clinical practice. A steering committee selected preliminary questions, which were reviewed and refined. Evidence was appraised using the United States Preventive Services Task Force (USPSTF) criteria for evidence level and degree of recommendation. Using a modified Delphi approach, consensus points were distributed to all collaborators and each collaborator voted on each point. If collaborators provided a decision of "disagree" or "abstain", they were invited to provide a rationale in a non-blinded fashion to the committee chair, who incorporated the respective comments and distributed revised versions to the committee until consensus was achieved. Results Sixteen questions were selected for guideline development. Questions that were addressed included type of injectable biologics and mechanism, evidence in treating chronic pain indications (eg, tendinopathy, muscular pathology, osteoarthritis, intervertebral disc disease, neuropathic pain), role in surgical augmentation, dosing, comparative efficacy between injectable biologics, peri-procedural practices to optimize therapeutic response and quality of injectate, federal regulations, and complications with mitigating strategies. Conclusion In well-selected individuals with certain chronic pain indications, use of injectable biologics may provide superior analgesia, functionality, and/or quality of life compared to conventional medical management or placebo. Future high-quality randomized clinical trials are warranted with implementation of minimum reporting standards, standardization of preparation protocols, investigation of dose-response associations, and comparative analysis between different injectable biologics.
Collapse
Affiliation(s)
- Ryan S D'Souza
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yeng F Her
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nasir Hussain
- Department of Anesthesiology, The Ohio State Wexner Medical Center, Columbus, OH, USA
| | - Jay Karri
- Departments of Orthopedic Surgery and Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael E Schatman
- Department of Anesthesiology, Perioperative Care, & Pain Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Christopher Lam
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Thomas Buchheit
- Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Brennan J Boettcher
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | | | - Scott G Pritzlaff
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Sacramento, CA, USA
| | | | - Shane A Shapiro
- Department of Orthopedic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Johana Klasova
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jay S Grider
- Department of Anesthesiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Ryan Hubbard
- Department of Sports Medicine, Anderson Orthopedic Clinic, Arlington, VA, USA
| | - Eliana Ege
- Department of Physical Medicine & Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Shelby Johnson
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Max H Epstein
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Eva Kubrova
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Mohamed Ehab Ramadan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Alexandra Michelle Moreira
- Department of Physical Medicine & Rehabilitation, University of Miami/Jackson Memorial Hospital, Miami, FL, USA
| | - Swarnima Vardhan
- Department of Internal Medicine, Yale New Haven Health - Bridgeport Hospital, Bridgeport, CT, USA
| | - Yashar Eshraghi
- Department of Anesthesiology & Critical Care Medicine, Ochsner Health System, New Orleans, LA, USA
| | - Saba Javed
- Department of Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Newaj M Abdullah
- Department of Anesthesiology, University of Utah, Salt Lake City, UT, USA
| | - Paul J Christo
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Sudhir Diwan
- Department of Pain Medicine, Advanced Spine on Park Avenue, New York City, NY, USA
| | | | - Dawood Sayed
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Timothy R Deer
- Department of Anesthesiology and Pain Medicine, West Virginia University School of Medicine, Charleston, WV, USA
| |
Collapse
|
6
|
Margulies BS, Loy JC, Thakur N, Sanz-Altamira P. Treating Multilevel Cervical Degenerative Disk Disease in a Patient With Stage IV Lung Cancer With Notable Comorbidities Using a Drug Eluting Biomaterial: A Case Report. J Am Acad Orthop Surg Glob Res Rev 2024; 8:01979360-202409000-00012. [PMID: 39312687 PMCID: PMC11421713 DOI: 10.5435/jaaosglobal-d-24-00051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/23/2024] [Accepted: 07/27/2024] [Indexed: 09/25/2024]
Abstract
A 64-year-old patient with stage IV non-small-cell lung carcinoma and several comorbidities, which include obesity and long-term smoking, was treated with N-allyl noroxymorphone eluting osteoinductive bone graft biomaterial. The patient had multilevel degenerative disk disease (DDD), which has a high rate of failure when osteoinductive bone grafts are not used. Infuse, the most widely administered osteoinductive bone graft, is contraindicated in the spine for patients with active tumor. As such, a novel drug eluting osteoinductive biomaterial was administered to this patient, for whom no other therapeutic options were available, to promote bone fusion in a three-level anterior cervical diskectomy and fusion as part of the Food and Drug Administration Expanded Access program. Despite patient comorbidities that are associated with poor bone physiology, confirmed radiographic fusion was achieved in all three cervical levels at 8 months.
Collapse
Affiliation(s)
- Bryan S Margulies
- From Zetagen Therapeutics, Syracuse, NY (Dr. Margulies, Mr. Loy, and Dr. Thakur); the Department of Pathology, College of Medicine, Upstate Medical University, Syracuse, NY (Dr. Margulies); the Department Biological Sciences, University of Notre Dame, Notre Dame, IN (Dr. Margulies); the Mobility Bone and Joint Institute, Andover, MA (Dr. Thakur); and the Dana-Farber Cancer Institute, Boston, MA (Dr. Sanz-Altamira)
| | | | | | | |
Collapse
|
7
|
Kuznetsova VS, Vasilyev AV, Bukharova TB, Nedorubova IA, Goldshtein DV, Popov VK, Kulakov AA. Application of BMP-2 and its gene delivery vehicles in dentistry. Saudi Dent J 2024; 36:855-862. [PMID: 38883899 PMCID: PMC11178965 DOI: 10.1016/j.sdentj.2024.03.015] [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: 11/14/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 06/18/2024] Open
Abstract
The restoration of bone defects resulting from tooth loss, periodontal disease, severe trauma, tumour resection and congenital malformations is a crucial task in dentistry and maxillofacial surgery. Growth factor- and gene-activated bone graft substitutes can be used instead of traditional materials to solve these problems. New materials will overcome the low efficacy and difficulties associated with the use of traditional bone substitutes in complex situations. One of the most well-studied active components for bone graft substitutes is bone morphogenetic protein-2 (BMP-2), which has strong osteoinductive properties. The aim of this review was to examine the use of BMP-2 protein and gene therapy for bone regeneration in the oral and maxillofacial region and to discuss its future use.
Collapse
Affiliation(s)
- Valeriya Sergeevna Kuznetsova
- Central Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
- Research Centre for Medical Genetics, Moscow, Russia
| | - Andrey Vyacheslavovich Vasilyev
- Central Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
- Research Centre for Medical Genetics, Moscow, Russia
| | | | | | | | - Vladimir Karpovich Popov
- Federal Scientific Research Centre "Crystallography and Photonics", Russian Academy of Sciences, Moscow, Russia
| | | |
Collapse
|
8
|
García-Lamas L, Lozano D, Jiménez-Díaz V, Bravo-Giménez B, Sánchez-Salcedo S, Jiménez-Holguín J, Abella M, Desco M, Vallet-Regi M, Cecilia-López D, Salinas AJ. Enriched mesoporous bioactive glass scaffolds as bone substitutes in critical diaphyseal bone defects in rabbits. Acta Biomater 2024; 180:104-114. [PMID: 38583750 DOI: 10.1016/j.actbio.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/23/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
In the field of orthopedic surgery, there is an increasing need for the development of bone replacement materials for the treatment of bone defects. One of the main focuses of biomaterials engineering are advanced bioceramics like mesoporous bioactive glasses (MBG´s). The present study compared the new bone formation after 12 weeks of implantation of MBG scaffolds with composition 82,5SiO2-10CaO-5P2O5-x 2.5SrO alone (MBGA), enriched with osteostatin, an osteoinductive peptide, (MBGO) or enriched with bone marrow aspirate (MBGB) in a long bone critical defect in radius bone of adult New Zealand rabbits. New bone formation from the MBG scaffold groups was compared to the gold standard defect filled with iliac crest autograft and to the unfilled defect. Radiographic follow-up was performed at 2, 6, and 12 weeks, and microCT and histologic examination were performed at 12 weeks. X-Ray study showed the highest bone formation scores in the group with the defect filled with autograft, followed by the MBGB group, in addition, the microCT study showed that bone within defect scores (BV/TV) were higher in the MBGO group. This difference could be explained by the higher density of newly formed bone in the osteostatin enriched MBG scaffold group. Therefore, MBG scaffold alone and enriched with osteostatin or bone marrow aspirate increase bone formation compared to defect unfilled, being higher in the osteostatin group. The present results showed the potential to treat critical bone defects by combining MBGs with osteogenic peptides such as osteostatin, with good prospects for translation into clinical practice. STATEMENT OF SIGNIFICANCE: Treatment of bone defects without the capacity for self-repair is a global problem in the field of Orthopedic Surgery, as evidenced by the fact that in the U.S alone it affects approximately 100,000 patients per year. The gold standard of treatment in these cases is the autograft, but its use has limitations both in the amount of graft to be obtained and in the morbidity produced in the donor site. In the field of materials engineering, there is a growing interest in the development of a bone substitute equivalent. Mesoporous bioactive glass (MBG´s) scaffolds with three-dimensional architecture have shown great potential for use as a bone substitutes. The osteostatin-enriched Sr-MBG used in this long bone defect in rabbit radius bone in vivo study showed an increase in bone formation close to autograft, which makes us think that it may be an option to consider as bone substitute.
Collapse
Affiliation(s)
- Lorena García-Lamas
- Servicio de Cirugía Ortopédica y Traumatología. Hospital Universitario 12 de Octubre, Madrid, España; Instituto de Investigación I+12, Madrid, España.
| | - Daniel Lozano
- Instituto de Investigación I+12, Madrid, España; Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, España
| | - Verónica Jiménez-Díaz
- Servicio de Cirugía Ortopédica y Traumatología. Hospital Universitario 12 de Octubre, Madrid, España; Instituto de Investigación I+12, Madrid, España
| | - Beatriz Bravo-Giménez
- Servicio de Cirugía Ortopédica y Traumatología. Hospital Universitario 12 de Octubre, Madrid, España; Instituto de Investigación I+12, Madrid, España
| | - Sandra Sánchez-Salcedo
- Instituto de Investigación I+12, Madrid, España; Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, España
| | - Javier Jiménez-Holguín
- Instituto de Investigación I+12, Madrid, España; Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, España
| | - Mónica Abella
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, España; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, España
| | - Manuel Desco
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, España; Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, España
| | - María Vallet-Regi
- Instituto de Investigación I+12, Madrid, España; Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, España
| | - David Cecilia-López
- Servicio de Cirugía Ortopédica y Traumatología. Hospital Universitario 12 de Octubre, Madrid, España; Instituto de Investigación I+12, Madrid, España
| | - Antonio Jesús Salinas
- Instituto de Investigación I+12, Madrid, España; Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, España.
| |
Collapse
|
9
|
Seok MC, Koo HW, Jeong JH, Ko MJ, Lee BJ. Bone Substitute Options for Spine Fusion in Patients With Spine Trauma-Part II: The Role of rhBMP. Korean J Neurotrauma 2024; 20:35-44. [PMID: 38576507 PMCID: PMC10990692 DOI: 10.13004/kjnt.2024.20.e13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/26/2023] [Accepted: 01/14/2024] [Indexed: 04/06/2024] Open
Abstract
In Part II, we focus on an important aspect of spine fusion in patients with spine trauma: the pivotal role of recombinant human bone morphogenetic protein-2 (rhBMP-2). Despite the influx of diverse techniques facilitated by technological advancements in spinal surgery, spinal fusion surgery remains widely used globally. The persistent challenge of spinal pseudarthrosis has driven extensive efforts to achieve clinically favorable fusion outcomes, with particular emphasis on the evolution of bone graft substitutes. Part II of this review aims to build upon the foundation laid out in Part I by providing a comprehensive summary of commonly utilized bone graft substitutes for spinal fusion in patients with spinal trauma. Additionally, it will delve into the latest advancements and insights regarding the application of rhBMP-2, offering an updated perspective on its role in enhancing the success of spinal fusion procedures.
Collapse
Affiliation(s)
- Min cheol Seok
- Department of Neurosurgery, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Hae-Won Koo
- Department of Neurosurgery, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Je Hoon Jeong
- Department of Neurosurgery, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Myeong Jin Ko
- Department of Neurosurgery, College of Medicine, Chung-Ang University Hospital, Seoul, Korea
| | - Byung-Jou Lee
- Department of Neurosurgery, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| |
Collapse
|
10
|
Raitio A, Saarinen AJ, Sinikumpu JJ, Helenius I. Biodegradable biomaterials in orthopedic surgery: A narrative review of the current evidence. Scand J Surg 2024; 113:62-70. [PMID: 37817459 DOI: 10.1177/14574969231200650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
BACKGROUND Biomaterials are routinely used in orthopedic surgery to fill bone defects, improve bone healing, and as degradable fixation material. A wide range of materials are currently in use, and the materials are chosen according to their bioactive properties. Osteoinductive materials stimulate bone healing by promoting osteogenesis. Osteoconductive materials facilitate bone growth on the surface of the material. Despite the many materials in use and an increasing number of published studies, randomized controlled trials on the subject are scarce. METHODS This review aims to summarize the history of biodegradable biomaterials and also the published level I evidence currently available on orthopedic biomaterials. RESULTS Most of the studies have been superiority trials with non-significant differences compared to conventional treatment options, confirming that several biomaterials are suitable treatment options for multiple indications including bone and/or tendon fixation, filling bone defects, and spinal fusion. Biomaterials help to avoid donor site complications associated with autogenous bone grafts and often eliminate the need for implant removal. However, the surgical technique may in some cases be more demanding than with conventional methods. Careful consideration of the pros and cons is therefore recommended in clinical practice. CONCLUSION Biodegradable biomaterials complement the range of available treatment options in several fields of orthopedic surgery. However, some biomaterials performed worse than expected and were not recommended for clinical use, emphasizing the need for high-quality randomized trials. It is also noteworthy that several trials included only a limited number of patients, rendering the interpretation of the results of these underpowered studies challenging.
Collapse
Affiliation(s)
- Arimatias Raitio
- Arimatias Raitio Department of Pediatric SurgeryUniversity of Turku and Turku University Hospital Turku Finland
- Turku University Hospital, Savitehtaankatu 5, 20520 Turku
| | - Antti J Saarinen
- Department of Orthopedics and Traumatology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Juha-Jaakko Sinikumpu
- Department of Pediatric Surgery and Orthopedics, Clinical Medicine Research Unit, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Ilkka Helenius
- Department of Pediatric Surgery, Orthopaedics and Traumatology, University of Turku and Turku University Hospital, Turku, Finland
- Department of Orthopedics and Traumatology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
11
|
Hong YR, Kim TH, Lee K, Lim JO, Oh CW. Bioactive Bone Substitute in a Rabbit Ulna Model: Preclinical Study. Tissue Eng Regen Med 2023; 20:1205-1217. [PMID: 37815697 PMCID: PMC10645865 DOI: 10.1007/s13770-023-00591-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Current therapies to effectively treat long-bone defects and extensive bone tissue loss remains limited. In this study, we created a new bone substitute by integrating advanced technologies such as structure patterning, controlled release of a bone growth factor and conjugation system for clinically effective bone regeneration. This novel bioactive bone substitute was evaluated for its safety and efficacy using a rabbit ulna model. METHODS A three dimensional bone patterned cylindrical structure with 1.5 cm in length and 5 mm in diameter was printed using poly(L-lactic acid)(PLLA) as a weight-bearing support and space-filling scaffold. And a bone morphogenetic protein 2 (BMP2) was employed to enhance bone regeneration, and coated to a 3D PLLA using alginate catechol and collagen to prolong the release kinetics. This novel bone substitute (BS)was evaluated for its physico-chemical and biological properties in vitro, and histological analysis and radiographical analysis such as X-ray, CT and micro-CT image analysis were performed to evaluate new bone formation in vivo. RESULTS The BS possesses an ideal shape and mechanically suitable proeperties for clinical use, with an easy-to-grab and break-resistant design at both ends, 80 ± 10 MPa of compression strength, and BMP2 release for two months. Histological analysis demonstrated the biocompability of BS with minimal inflammation and immune response, and X-ray, CT and micro-CT demonstrated effective new bone formation in rabbit ulna defect model. CONCLUSION The preclinical study of a novel bioactive bone substitute has shown its safe and effective properties in an animal model suggesting its clinical potential.
Collapse
Affiliation(s)
- Yu Ri Hong
- Bio-Medical Research Institute, Kyungpook National University Hospital, Daegu, 41940, Republic of Korea
- Joint Institute for Regenerative Medicine, Kyungpook National University, Daegu, 41940, Republic of Korea
| | - Tae-Ho Kim
- Bio-Medical Research Institute, Kyungpook National University Hospital, Daegu, 41940, Republic of Korea
- Joint Institute for Regenerative Medicine, Kyungpook National University, Daegu, 41940, Republic of Korea
| | - Kyueui Lee
- Department of Chemistry and Green-Nano Materials Research Center, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jeong Ok Lim
- Bio-Medical Research Institute, Kyungpook National University Hospital, Daegu, 41940, Republic of Korea.
- School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Joint Institute for Regenerative Medicine, Kyungpook National University, Daegu, 41940, Republic of Korea.
| | - Chang-Wug Oh
- School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Orthopedic Surgery, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea.
| |
Collapse
|
12
|
Burioli EAV, Hammel M, Vignal E, Vidal-Dupiol J, Mitta G, Thomas F, Bierne N, Destoumieux-Garzón D, Charrière GM. Transcriptomics of mussel transmissible cancer MtrBTN2 suggests accumulation of multiple cancer traits and oncogenic pathways shared among bilaterians. Open Biol 2023; 13:230259. [PMID: 37816387 PMCID: PMC10564563 DOI: 10.1098/rsob.230259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
Transmissible cancer cell lines are rare biological entities giving rise to diseases at the crossroads of cancer and parasitic diseases. These malignant cells have acquired the amazing capacity to spread from host to host. They have been described only in dogs, Tasmanian devils and marine bivalves. The Mytilus trossulus bivalve transmissible neoplasia 2 (MtrBTN2) lineage has even acquired the capacity to spread inter-specifically between marine mussels of the Mytilus edulis complex worldwide. To identify the oncogenic processes underpinning the biology of these atypical cancers we performed transcriptomics of MtrBTN2 cells. Differential expression, enrichment, protein-protein interaction network, and targeted analyses were used. Overall, our results suggest the accumulation of multiple cancerous traits that may be linked to the long-term evolution of MtrBTN2. We also highlight that vertebrate and lophotrochozoan cancers could share a large panel of common drivers, which supports the hypothesis of an ancient origin of oncogenic processes in bilaterians.
Collapse
Affiliation(s)
- E A V Burioli
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - M Hammel
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
- ISEM, Univ Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - E Vignal
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - J Vidal-Dupiol
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - G Mitta
- IFREMER, UMR 241 Écosystèmes Insulaires Océaniens, Labex Corail, Centre Ifremer du Pacifique, Tahiti, Polynésie française
| | - F Thomas
- CREEC/CANECEV (CREES), MIVEGEC, Unité Mixte de Recherches, IRD 224-CNRS 5290-Université de Montpellier, Montpellier, France
| | - N Bierne
- ISEM, Univ Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - D Destoumieux-Garzón
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - G M Charrière
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| |
Collapse
|
13
|
Patel D, Tatum SA. Bone Graft Substitutes and Enhancement in Craniomaxillofacial Surgery. Facial Plast Surg 2023; 39:556-563. [PMID: 37473765 DOI: 10.1055/s-0043-1770962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023] Open
Abstract
Critical-sized bone defects are a reconstructive challenge, particularly in the craniomaxillofacial (CMF) skeleton. The "gold standard" of autologous bone grafting has been the work horse of reconstruction in both congenital and acquired defects of CMF skeleton. Autologous bone has the proper balance of the protein (or organic) matrix and mineral components with no immune response. Organic and mineral adjuncts exist that offer varying degrees of osteogenic, osteoconductive, osteoinductive, and osteostimulative properties needed for treatment of critical-sized defects. In this review, we discuss the various mostly organic and mostly mineral bone graft substitutes available for autologous bone grafting. Primarily organic bone graft substitutes/enhancers, including bone morphogenic protein, platelet-rich plasma, and other growth factors, have been utilized to support de novo bone growth in setting of critical-sized bone defects. Primarily mineral options, including various calcium salt formulation (calcium sulfate/phosphate/apatite) and bioactive glasses have been long utilized for their similar composition to bone. Yet, a bone graft substitute that can supplant autologous bone grafting is still elusive. However, case-specific utilization of bone graft substitutes offers a wider array of reconstructive options.
Collapse
Affiliation(s)
- Dhruv Patel
- Department of Otolaryngology, SUNY Upstate Medical University, Syracuse, New York
| | - Sherard A Tatum
- Department of Otolaryngology and Pediatrics, SUNY Upstate Medical University, Syracuse, New York
| |
Collapse
|
14
|
Munim MA, Berlinberg E, Federico VP, Nolte MT, Prabhu M, Pawlowski H, Patel KS, Colman MW. Usage Trends and Safety Profile of Recombinant Human Bone Morphogenetic Protein-2 for Spinal Column Tumor Surgery: A National Matched Cohort Analysis. Global Spine J 2023:21925682231194248. [PMID: 37542521 DOI: 10.1177/21925682231194248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/07/2023] Open
Abstract
STUDY DESIGN Retrospective Cohort Analysis. OBJECTIVE The purpose of this study is to investigate national rates of rhBMP-2 utilization in spinal tumor surgery and examine its association with postoperative complications, revisions, and carcinogenicity. METHODS All patients diagnosed with primary or metastatic spinal tumors with subsequent surgical intervention involving a spinal fusion procedure were identified in PearlDiver. Patients were 1:1 matched into 2 cohorts according to rhBMP-2 usage. Postoperative complications and revisions were examined at 1 month, 3 months, 6 months, and 1 year after fusion. New cancer incidence following spinal tumor surgery was assessed until 5 years postoperatively. RESULTS A total of 11,198 patients underwent fusion surgery after resection of spinal tumors between 2005 and 2020, with 909 cases reporting the use of rhBMP-2 (8.1%). An annualized analysis revealed that the proportion of spine tumor fusion procedures utilizing rhBMP-2 has been significantly decreasing (R2 = .859, P < .001), with the most recent annual utilization rate at 1.1%. At least 3 months after surgery, significantly increased incidences of surgical site (11.4% vs 3.3%, P = .03) and systemic infections (8.1% vs 1.6%, P = .02) were observed in patients who underwent fusion with rhBMP-2. Across all time points, no significant differences were observed in survival, implant removal, revision rates, or new cancer diagnoses. CONCLUSION This analysis demonstrated significantly declining national utilization rates. Spinal tumor cases utilizing rhBMP-2 sustained greater rates of surgical site and systemic infections. rhBMP-2 usage did not significantly reduce the risk of mortality, implant failure, or reoperation.
Collapse
Affiliation(s)
- Mohammed A Munim
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Elyse Berlinberg
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Vincent P Federico
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Michael T Nolte
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Michael Prabhu
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Hanna Pawlowski
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Karan S Patel
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Matthew W Colman
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
15
|
Katrilaka C, Karipidou N, Petrou N, Manglaris C, Katrilakas G, Tzavellas AN, Pitou M, Tsiridis EE, Choli-Papadopoulou T, Aggeli A. Freeze-Drying Process for the Fabrication of Collagen-Based Sponges as Medical Devices in Biomedical Engineering. MATERIALS (BASEL, SWITZERLAND) 2023; 16:4425. [PMID: 37374608 DOI: 10.3390/ma16124425] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
This paper presents a systematic review of a key sector of the much promising and rapidly evolving field of biomedical engineering, specifically on the fabrication of three-dimensional open, porous collagen-based medical devices, using the prominent freeze-drying process. Collagen and its derivatives are the most popular biopolymers in this field, as they constitute the main components of the extracellular matrix, and therefore exhibit desirable properties, such as biocompatibility and biodegradability, for in vivo applications. For this reason, freeze-dried collagen-based sponges with a wide variety of attributes can be produced and have already led to a wide range of successful commercial medical devices, chiefly for dental, orthopedic, hemostatic, and neuronal applications. However, collagen sponges display some vulnerabilities in other key properties, such as low mechanical strength and poor control of their internal architecture, and therefore many studies focus on the settlement of these defects, either by tampering with the steps of the freeze-drying process or by combining collagen with other additives. Furthermore, freeze drying is still considered a high-cost and time-consuming process that is often used in a non-optimized manner. By applying an interdisciplinary approach and combining advances in other technological fields, such as in statistical analysis, implementing the Design of Experiments, and Artificial Intelligence, the opportunity arises to further evolve this process in a sustainable and strategic manner, and optimize the resulting products as well as create new opportunities in this field.
Collapse
Affiliation(s)
- Chrysoula Katrilaka
- Department of Chemical Engineering, School of Engineering, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Niki Karipidou
- Department of Chemical Engineering, School of Engineering, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Nestor Petrou
- Department of Chemical Engineering, School of Engineering, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Chris Manglaris
- Department of Chemical Engineering, School of Engineering, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - George Katrilakas
- Department of Chemical Engineering, School of Engineering, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Anastasios Nektarios Tzavellas
- 3rd Department of Orthopedics, School of Medicine, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Maria Pitou
- School of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Eleftherios E Tsiridis
- 3rd Department of Orthopedics, School of Medicine, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | | | - Amalia Aggeli
- Department of Chemical Engineering, School of Engineering, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
16
|
Atasoy-Zeybek A, Coenen MJ, Hawse GP, Logeart-Avramoglou D, Evans CH, De La Vega RE. Efficient autocrine and paracrine signaling explain the osteogenic superiority of transgenic BMP-2 over rhBMP-2. Mol Ther Methods Clin Dev 2023; 29:350-363. [PMID: 37214314 PMCID: PMC10196773 DOI: 10.1016/j.omtm.2023.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Bone morphogenetic protein-2 (BMP-2) is an osteogenic protein used clinically to enhance bone healing. However, it must be applied in very high doses, causing adverse side effects and increasing costs while providing only incremental benefit. Preclinical models of bone healing using gene transfer to deliver BMP-2 suggest that transgenic BMP-2 is much more osteogenic than rhBMP-2. Using a reporter mesenchymal cell line, we found transgenic human BMP-2 cDNA to be at least 100-fold more effective than rhBMP-2 in signaling. Moreover, a substantial portion of the BMP-2 produced by the transduced cells remained cell associated. Signaling by transgenic BMP-2 occurred via binding to the type I receptor, activating the associated kinase and generating phospho-smads. Signaling was partially resistant to noggin, an important extracellular inhibitor of BMP-2, possibly because nascent BMP-2 binds to its cell surface receptor during secretion and thus signals in a protected peri-cellular environment. Although the amounts of BMP-2 secreted by the transduced cells were too low to affect distant cells, transduced cells were able to induce signaling in a paracrine fashion that required close proximity of the cells, possibly cell-to-cell contact. The greater osteogenic potency of transgenic BMP-2 was confirmed with human bone marrow stromal cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Rodolfo E. De La Vega
- Mayo Clinic, Rochester, MN, USA
- cBITE, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
17
|
Kupka JR, Sagheb K, Al-Nawas B, Schiegnitz E. The Sympathetic Nervous System in Dental Implantology. J Clin Med 2023; 12:jcm12082907. [PMID: 37109243 PMCID: PMC10143978 DOI: 10.3390/jcm12082907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
The sympathetic nervous system plays a vital role in various regulatory mechanisms. These include the well-known fight-or-flight response but also, for example, the processing of external stressors. In addition to many other tissues, the sympathetic nervous system influences bone metabolism. This effect could be highly relevant concerning osseointegration, which is responsible for the long-term success of dental implants. Accordingly, this review aims to summarize the current literature on this topic and to reveal future research perspectives. One in vitro study showed differences in mRNA expression of adrenoceptors cultured on implant surfaces. In vivo, sympathectomy impaired osseointegration in mice, while electrical stimulation of the sympathetic nerves promoted it. As expected, the beta-blocker propranolol improves histological implant parameters and micro-CT measurements. Overall, the present data are considered heterogeneous. However, the available publications reveal the potential for future research and development in dental implantology, which helps to introduce new therapeutic strategies and identify risk factors for dental implant failure.
Collapse
Affiliation(s)
- Johannes Raphael Kupka
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, 55131 Mainz, Germany
| | - Keyvan Sagheb
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, 55131 Mainz, Germany
| | - Bilal Al-Nawas
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, 55131 Mainz, Germany
| | - Eik Schiegnitz
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, 55131 Mainz, Germany
| |
Collapse
|
18
|
Pinto N, Klein Y, David E, Polak D, Steinberg D, Mizrahi G, Khoury Y, Barenholz Y, Chaushu S. Resolvin D1 improves allograft osteointegration and directly enhances osteoblasts differentiation. Front Immunol 2023; 14:1086930. [PMID: 36923414 PMCID: PMC10008843 DOI: 10.3389/fimmu.2023.1086930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/03/2023] [Indexed: 03/02/2023] Open
Abstract
Introduction Allografts are the most common bone grafts for repairing osseous defects. However, their use is associated with an increased risk for infections, donor disease transmission and osteointegration deficiency. Resolvin D1 (RvD1) is an endogenous lipid with a scientifically proven pivotal role in inflammation resolution and osteoclastogenesis inhibition. Yet, its biological relevance as a potential bone regenerative drug has been scarcely studied. Here, we aim to investigate the RvD1 effect on allograft osteointegration in the alveolar bone regeneration (ABR) murine model. Methods ABR model consisted of osseous defects that were generated by the extraction of the maxillary first molar in C57BL/6 mice. The sockets were filled with allograft and analyzed via RNA sequencing. Then they were locally injected with either RvD1 or saline via single or repeated administrations. The mice were sacrificed 2W after the procedure, and regenerated sites were analyzed using µCT and histology. First, MC3T3-E1 preosteoblasts were plated with IL-17 pro-inflammatory medium, and RANKL/OPG ratio was measured. Secondly, the MC3T3-E1 were cultured w/o RvD1, for 3W. Osteoblasts' markers were evaluated in different days, using qRT-PCR and Alizarin Red staining for calcified matrix. Results In vivo, neither allograft alone nor single RvD1 administration promote bone regeneration in comparison to the control of spontaneous healing and even triggered an elevation in NR1D1 and IL1RL1 expression, markers associated with inflammation and inhibition of bone cell differentiation. However, repeated RvD1 treatment increased bone content by 135.92% ± 45.98% compared to its specific control, repeated sham, and by 39.12% ± 26.3% when compared to the spontaneous healing control group (n=7/group). Histologically, repeated RvD1 reduced the number of TRAP-positive cells, and enhanced allograft osteointegration with new bone formation. In vitro, RvD1 rescued OPG expression and decreased RANKL/OPG ratio in IL-17 pro-inflammatory conditions. Furthermore, RvD1 increased the expression of RUNX2, OSX, BSP and OC/BGLAP2 and the mineralized extracellular matrix during MC3T3-E1 osteoblasts differentiation. Conclusions Repeated administrations of RvD1 promote bone regeneration via a dual mechanism: directly, via enhancement of osteoblasts' differentiation and indirectly, through reduction of osteoclastogenesis and RANKL/OPG ratio. This suggests that RvD1 may be a potential therapeutic bioagent for osseous regeneration following allograft implantation.
Collapse
Affiliation(s)
- Noy Pinto
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yehuda Klein
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Department of Orthodontics, Hadassah Medical Center, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eilon David
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - David Polak
- Department of Periodontics, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Daniel Steinberg
- The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-Medical Research, Israel-Canada (IMRIC), Jerusalem, Israel.,Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gilad Mizrahi
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Department of Orthodontics, Hadassah Medical Center, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yasmin Khoury
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yechezkel Barenholz
- Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Stella Chaushu
- Department of Orthodontics, Hadassah Medical Center, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
19
|
BMP2 as a promising anticancer approach: functions and molecular mechanisms. Invest New Drugs 2022; 40:1322-1332. [PMID: 36040572 DOI: 10.1007/s10637-022-01298-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/22/2022] [Indexed: 10/14/2022]
Abstract
Bone morphogenetic protein 2 (BMP2), a pluripotent factor, is a member of the transforming growth factor-beta (TGF-β) superfamily and is implicated in embryonic development and postnatal homeostasis in tissues and organs. Experimental research in the contexts of physiology and pathology has indicated that BMP2 can induce macrophages to differentiate into osteoclasts and accelerate the osteolytic mechanism, aggravating cancer cell bone metastasis. Emerging studies have stressed the potent regulatory effect of BMP2 in cancer cell differentiation, proliferation, survival, and apoptosis. Complicated signaling networks involving multiple regulatory proteins imply the significant biological functions of BMP2 in cancer. In this review, we comprehensively summarized and discussed the current evidence related to the modulation of BMP2 in tumorigenesis and development, including evidence related to the roles and molecular mechanisms of BMP2 in regulating cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT), cancer angiogenesis and the tumor microenvironment (TME). All these findings suggest that BMP2 may be an effective therapeutic target for cancer and a new marker for assessing treatment efficacy.
Collapse
|
20
|
Discovery of Some Heterocyclic Molecules as Bone Morphogenetic Protein 2 (BMP-2)-Inducible Kinase Inhibitors: Virtual Screening, ADME Properties, and Molecular Docking Simulations. Molecules 2022; 27:molecules27175571. [PMID: 36080338 PMCID: PMC9457949 DOI: 10.3390/molecules27175571] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/13/2022] [Accepted: 08/18/2022] [Indexed: 12/02/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are growth factors that have a vital role in the production of bone, cartilage, ligaments, and tendons. Tumors’ upregulation of bone morphogenetic proteins (BMPs) and their receptors are key features of cancer progression. Regulation of the BMP kinase system is a new promising strategy for the development of anti-cancer drugs. In this work, based on a careful literature study, a library of benzothiophene and benzofuran derivatives was subjected to different computational techniques to study the effect of chemical structure changes on the ability of these two scaffolds to target BMP-2 inducible kinase, and to reach promising candidates with proposed activity against BMP-2 inducible kinase. The results of screening against Lipinski’s and Veber’s Rules produced twenty-one outside eighty-four compounds having drug-like molecular nature. Computational ADMET studies favored ten compounds (11, 26, 27, 29, 30, 31, 34, 35, 65, and 72) with good pharmacokinetic profile. Computational toxicity studies excluded compound 34 to elect nine compounds for molecular docking studies which displayed eight compounds (26, 27, 29, 30, 31, 35, 65, and 72) as promising BMP-2 inducible kinase inhibitors. The nine fascinating compounds will be subjected to extensive screening against serine/threonine kinases to explore their potential against these critical proteins. These promising candidates based on benzothiophene and benzofuran scaffolds deserve further clinical investigation as BMP-2 kinase inhibitors for the treatment of cancer.
Collapse
|
21
|
Chen Z, Yuan L, Li X, Yu J, Xu Z. BMP2 inhibits cell proliferation by downregulating EZH2 in gastric cancer. Cell Cycle 2022; 21:2298-2308. [PMID: 35856444 DOI: 10.1080/15384101.2022.2092819] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Gastric cancer is among the most common gastrointestinal malignancies. Recent studies have suggested that bone morphogenetic protein-2 (BMP2) is related to the development and progression of various cancers. Meanwhile, evidence suggests that BMP2 might lead to epigenetic changes in gastric cancer. Thus, we investigated whether BMP2 plays a role in the development of gastric cancer via epigenetic regulation. Cell viability, colony formation, and cell cycle assays were performed to assess the effect of recombinant human BMP2 (rhBMP2) in gastric cancer cells. LDN-193189 and Noggins were used as antagonists of the canonical BMP-SMAD signaling pathway. The protein levels were determined using a western blot analysis. Lentiviral vectors with EZH2 shRNA or EZH2 overexpression were used to mediate the role of EZH2 and the relationship between BMP2 and EZH2 in gastric cancer. We found that rhBMP2 inhibits cell proliferation by arresting the cell cycle in HGC-27 and SNU-216 gastric cancer cells. Neither LDN-193189 nor Noggins, antagonists of the canonical BMP-SMAD signaling pathway, can reverse the effect of rhBMP2 on gastric cancer. Molecularly, rhBMP2 downregulates the expression of EZH2 and H3K27me3, leading to increases in P16 and P21 and decreases in CDK2, CDK4, and CDK6. Altogether, in this study, we demonstrate that BMP2 serves as a tumor suppressor in gastric cancer cells by downregulating EZH2 and H3K27me3 through the non-SMAD BMP pathway, suggesting that BMP2 might be a new therapeutic target for gastric cancer treatment. Abbreviations: BMP: bone morphogenetic protein; TGF-β: transforming growth factor-beta; EZH2: enhancer of zeste homolog 2; H3K27me3: trimethylation histone H3 lysine 27; HRECs: human retinal endothelial cells; PcG: polycomb group; PRC: polycomb repressive complexes.
Collapse
Affiliation(s)
- Zilu Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liyue Yuan
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaopeng Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Junhui Yu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhengshui Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Hixon KR, Katz DB, McKenzie JA, Miller AN, Guilak F, Silva MJ. Cryogel Scaffold-Mediated Delivery of Adipose-Derived Stem Cells Promotes Healing in Murine Model of Atrophic Non-Union. Front Bioeng Biotechnol 2022; 10:851904. [PMID: 35600896 PMCID: PMC9117654 DOI: 10.3389/fbioe.2022.851904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/23/2022] [Indexed: 01/08/2023] Open
Abstract
Non-union is defined as the permanent failure of a bone to heal and occurs clinically in 5% of fractures. Atrophic non-unions, characterized by absent/minimal callus formation, are poorly understood and difficult to treat. We recently demonstrated a novel murine model of atrophic non-union in the 3.6Col1A1-tk (Col1-tk) mouse, wherein dosing with the nucleoside analog ganciclovir (GCV) was used to deplete proliferating osteoprogenitor cells, leading to a radiographic and biomechanical non-union after the mid-shaft femur fracture. Using this Col1-tk atrophic non-union model, we hypothesized that the scaffold-mediated lentiviral delivery of doxycycline-inducible BMP-2 transgenes would induce osteogenesis at the fracture site. Cryogel scaffolds were used as a vehicle for GFP+ and BMP-2+ cell delivery to the site of non-union. Cryogel scaffolds were biofabricated through the cross-linking of a chitosan-gelatin polymer solution at subzero temperatures, which results in a macroporous, spongy structure that may be advantageous for a bone regeneration application. Murine adipose-derived stem cells were seeded onto the cryogel scaffolds, where they underwent lentiviral transduction. Following the establishment of atrophic non-unions in the femurs of Col1-tk mice (4 weeks post-fracture), transduced, seeded scaffolds were surgically placed around the site of non-union, and the animals were given doxycycline water to induce BMP-2 production. Controls included GFP+ cells on the cryogel scaffolds, acellular scaffolds, and sham (no scaffold). Weekly radiographs were taken, and endpoint analysis included micro-CT and histological staining. After 2 weeks of implantation, the BMP-2+ scaffolds were infiltrated with cartilage and woven bone at the non-union site, while GFP+ scaffolds had woven bone formation. Later, timepoints of 8 weeks had woven bone and vessel formation within the BMP-2+ and GFP + scaffolds with cortical bridging of the original fracture site in both groups. Overall, the cell-seeded cryogels promoted osseous healing. However, while the addition of BMP-2 promoted the endochondral ossification, it may provide a slower route to healing. This proof-of-concept study demonstrates the potential for cellularized cryogel scaffolds to enhance the healing of non-unions.
Collapse
Affiliation(s)
- Katherine R. Hixon
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Dakota B. Katz
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
- Center of Regenerative Medicine, Washington University, St. Louis, MO, United States
- Shriners Hospitals for Children—St. Louis, St. Louis, MO, United States
| | - Jennifer A. McKenzie
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
| | - Anna N. Miller
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
- Center of Regenerative Medicine, Washington University, St. Louis, MO, United States
- Shriners Hospitals for Children—St. Louis, St. Louis, MO, United States
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
- Center of Regenerative Medicine, Washington University, St. Louis, MO, United States
| |
Collapse
|
23
|
Abstract
Purpose of Review Despite the continued growth of spine fusion procedures, the ideal material for bone regeneration remains unclear. Current bone graft substitutes and extenders in use such as exogenous BMP-2 or demineralized bone matrix and hydroxyapatite either have serious complications associated with use or lead to clinically significant rates of non-union. The introduction of nanotechnology and 3D printing to regenerative medicine facilitates the development of safer and more efficacious bone regenerative scaffolds that present solutions to these problems. Many researchers in orthopedics recognize the importance of lowering the dose of recombinant growth factors like BMP-2 to avoid the complications associated with its normal required supraphysiologic dosing to achieve high rates of fusion in spine surgery. Recent Findings Recent iterations of bioactive scaffolds have moved towards peptide amphiphiles that bind endogenous osteoinductive growth factor sources at the site of implantation. These molecules have been shown to provide a highly fluid, natural mimetic of natural extracellular matrix to achieve 100% fusion rates at 10–100 times lower doses of BMP-2 relative to controls in pre-clinical animal posterolateral fusion models. Alternative approaches to bone regeneration include the combination of existing natural growth factor sources like human bone combined with bioactive, biocompatible components like hydroxyapatite using 3D-printing technologies. Their elastomeric, 3D-printed scaffolds demonstrate an optimal safety profile and high rates of fusion (~92%) in the rat posterolateral fusion model. Summary Bioactive peptide amphiphiles and developments in 3D printing offer the promising future of a recombinant growth factor- free bone graft substitute with similar efficacy but improved safety profiles compared to existing bone graft substitutes.
Collapse
|
24
|
Tang C, Liang D, Qiu Y, Zhu J, Tang G. Omentin‑1 induces osteoblast viability and differentiation via the TGF‑β/Smad signaling pathway in osteoporosis. Mol Med Rep 2022; 25:132. [PMID: 35179221 PMCID: PMC8867465 DOI: 10.3892/mmr.2022.12648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 07/13/2021] [Indexed: 11/25/2022] Open
Abstract
Osteoporosis is a bone-related disease that results from impaired bone formation and excessive bone resorption. The potential value of adipokines has been investigated previously, due to their influence on osteogenesis. However, the osteogenic effects induced by omentin-1 remain unclear. The aim of the present study was to determine the regulatory effects of omentin-1 on osteoblast viability and differentiation, as well as to explore the underlying molecular mechanism. The present study investigated the effects of omentin-1 on the viability and differentiation of mouse pre-osteoblast cells (MC3T3-E1) using quantitative and qualitative measures. A Cell Counting Kit-8 assay was used to assess the viability of MC3T3-E1 cells following treatment with different doses of omentin-1. Omentin-1 and bone morphogenetic protein (BMP) inhibitor were added to osteogenic induction mediums in different ways to assess their effect. The alkaline phosphatase (ALP) activity and Alizarin Red S (ARS) staining of MC3T3-E1 cells treated with omentin-1 and/or BMP inhibitor were used to examine the effects of omentin-1 on differentiation and mineralization. Western blotting was used to further explore its potential mechanism, and to study the role of omentin-1 on the viability and differentiation of osteoblasts. The results showed that omentin-1 altered the viability of MC3T3-E1 cells in a dose-dependent manner. Omentin-1 treatment significantly increased the expression of members of the TGF-β/Smad signaling pathway. In the omentin-1 group, the ALP activity of the MC3T3-E1 cells was increased, and the ARS staining area was also increased. The mRNA and protein expression levels of BMP2, Runt-related transcription factor 2, collagen1, osteopontin, osteocalcin and osterix in the omentin-1 group were also significantly upregulated. All these effects were reversed following treatment with SIS3 HCl. These results demonstrated that omentin-1 can significantly promote osteoblast viability and differentiation via the TGF-β/Smad signaling pathway, thereby promoting bone formation and preventing osteoporosis.
Collapse
Affiliation(s)
- Cuisong Tang
- Department of Radiology, Clinical Medical College of Shanghai Tenth People's Hospital of Nanjing Medical University, Shanghai 200072, P.R. China
| | - Dengpan Liang
- Department of Cardiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Yuyou Qiu
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jingqi Zhu
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Guangyu Tang
- Department of Radiology, Clinical Medical College of Shanghai Tenth People's Hospital of Nanjing Medical University, Shanghai 200072, P.R. China
| |
Collapse
|
25
|
Liang W, Dong Y, Shen H, Shao R, Wu X, Huang X, Sun B, Zeng B, Zhang S, Xu F. Materials science and design principles of therapeutic materials in orthopedic and bone tissue engineering. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Wenqing Liang
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University Zhoushan China
| | - Yongqiang Dong
- Department of Orthopedics Xinchang People's Hospital Shaoxing China
| | - Hailiang Shen
- Department of Orthopedics Affiliated Hospital of Shaoxing University Shaoxing China
| | - Ruyi Shao
- Department of Orthopedics Zhuji People's Hospital Shaoxing China
| | - Xudong Wu
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University Zhoushan China
| | - Xiaogang Huang
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University Zhoushan China
| | - Bin Sun
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University Zhoushan China
| | - Bin Zeng
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University Zhoushan China
| | - Songou Zhang
- College of Medicine Shaoxing University Shaoxing China
| | - Fangming Xu
- Department of Orthopedics Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University Zhoushan China
| |
Collapse
|
26
|
Gong Y, Wang Y, Zhang Y, Wang L, Wan L, Zu Y, Li C, Wang X, Cui ZK. Paracrine Effects of Recombinant Human Adiponectin Promote Bone Regeneration. Front Cell Dev Biol 2021; 9:762335. [PMID: 34790669 PMCID: PMC8591230 DOI: 10.3389/fcell.2021.762335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 09/27/2021] [Indexed: 01/15/2023] Open
Abstract
Bone regeneration is a delicate physiological process. Non-union and delayed fracture healing remains a great challenge in clinical practice nowadays. Bone and fat hold a close relationship to remain balanced through hormones and cytokines. Adiponectin is a well-known protein to maintain the hemostasis, which may be an interesting target for fracture healing. Herein, we provided a facile and efficient method to obtain high-purity and high-yield recombinant human adiponectin (ADPN). The biocompatibility and the pharmaceutical behaviors were evaluated in Sprague–Dawley rats. The paracrine effects of adiponectin on bone fracture healing were investigated with a rat tibia fracture model via intrabone injection. Significantly accelerated bone healing was observed in the medulla injection group, indicating the paracrine effects of adiponectin could be potentially utilized for clinical treatments. The underlying mechanism was primarily assessed, and the expression of osteogenic markers, including bone morphogenic protein 2, alkaline phosphatase, and osteocalcin, along with adiponectin receptor 1 (AdipoR1), was markedly increased at the fracture site. The increased bone healing of ADPN treatment may result from both enhanced osteogenic proliferation as well as differentiation. Cell experiments confirmed that the expression of osteogenesis markers increased significantly in ADPN treatment groups, while it decreased when the expression of AdipoR1 was knocked down by siRNA. Our study provided a feasible and efficacious way for bone fracture treatment with local administration of ADPN, which could be rapidly translated into the clinics.
Collapse
Affiliation(s)
- Yanping Gong
- Department of Endocrinology, The Second Medical Center, National Clinical Research Center for Geriatric Disease, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yang Wang
- Department of Endocrinology, The Second Medical Center, National Clinical Research Center for Geriatric Disease, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yiqing Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Liangchen Wang
- Department of Endocrinology, The Second Medical Center, National Clinical Research Center for Geriatric Disease, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lijuan Wan
- Department of Endocrinology, The Second Medical Center, National Clinical Research Center for Geriatric Disease, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yuan Zu
- Department of Endocrinology, The Second Medical Center, National Clinical Research Center for Geriatric Disease, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Chunlin Li
- Department of Endocrinology, The Second Medical Center, National Clinical Research Center for Geriatric Disease, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xin Wang
- Institute of Orthopedics, The First Medical Center, The People's Liberation Army General Hospital, Beijing, China
| | - Zhong-Kai Cui
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Bioland Laboratory, Guangzhou, China
| |
Collapse
|
27
|
Body AM, Plummer ZJ, Krueger BM, Virojanapa J, Nasser R, Cheng JS, Prestigiacomo CJ. Retrograde ejaculation following anterior lumbar surgery: a systematic review and pooled analysis. J Neurosurg Spine 2021; 35:427-436. [PMID: 34271542 DOI: 10.3171/2020.12.spine201101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/21/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The present systematic review and pooled analysis aims to assess the incidence and risk factors for the development of retrograde ejaculation (RE) following first-time open anterior lumbar surgery. METHODS A systematic MEDLINE review via PubMed was performed, identifying 130 clinical papers relating to the topic. Eighteen publications were selected according to predetermined inclusion and exclusion criteria and were used to determine the incidence of RE. Only the publications that provided data on surgical risk factors present specifically in the men in the study were included in the analysis of risk factors. RESULTS Of the 2503 men included, there were 57 reported events of RE (2.3%). Of the cases for which long-term data were provided, 45.8% had resolved by final follow-up. There was a statistically significant increased risk associated with a transperitoneal as opposed to a retroperitoneal approach (8.6% vs 3.2%), as well as with the use of recombinant human bone morphogenetic protein-2 (rhBMP-2) in anterior lumbar interbody fusion (ALIF) as opposed to ALIF with bone graft or arthroplasty in controls (5.0% vs 1.8%). However, when excluding from analysis the patients operated on prior to the FDA's 2008 warning that commented on the drug's neuroinflammatory properties, there was no significant difference in rates of RE in patients receiving rhBMP-2 versus the control group (2.4% vs 2.5%). There was no significant difference in risk based on single- versus multilevel procedure or on ALIF versus arthroplasty. CONCLUSIONS In a pooled analysis of currently published data on men undergoing first-time open anterior lumbar surgery, this study found an overall incidence of RE of 2.3%. Nearly half of these patients recovered, reporting resolution of symptoms anywhere between 3 months and 48 months. Analysis of risk factors was limited by a paucity of published literature segregating data by sex. However, there was an increased risk associated with rhBMP-2 only when including data collected prior to the FDA warning on its detrimental properties. The authors therefore posit that the risk of RE is probably overestimated in the literature, given that the vast majority of the data available were collected prior to this warning and given the subsequent implementation of precautions when handling rhBMP-2.
Collapse
|
28
|
Li Z, Xiang S, Lin Z, Li EN, Yagi H, Cao G, Yocum L, Li L, Hao T, Bruce KK, Fritch MR, Hu H, Wang B, Alexander PG, Khor KA, Tuan RS, Lin H. Graphene oxide-functionalized nanocomposites promote osteogenesis of human mesenchymal stem cells via enhancement of BMP-SMAD1/5 signaling pathway. Biomaterials 2021; 277:121082. [PMID: 34464823 DOI: 10.1016/j.biomaterials.2021.121082] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/07/2021] [Accepted: 08/21/2021] [Indexed: 12/28/2022]
Abstract
Biomaterials that can harness the intrinsic osteogenic potential of stem cells offer a promising strategy to accelerate bone regeneration and repair. Previously, we had used methacrylated gelatin (GelMA)-based scaffolds to achieve bone formation from human mesenchymal stem cells (hMSCs). In this study, we aimed to further enhance hMSC osteogenesis by incorporating graphene oxide (GO)-based nanosheets into GelMA. In vitro results showed high viability and metabolic activities in hMSCs encapsulated in the newly developed nanocomposites. Incorporation of GO markedly increased mineralization within hMSC-laden constructs, which was further increased by replacing GO with silica-coated graphene oxide (SiGO). Mechanistic analysis revealed that the nanosheet enhanced the production, retention, and biological activity of endogenous bone morphogenetic proteins (BMPs), resulting in robust osteogenesis in the absence of exogenous osteoinductive growth factors. Specifically, the osteoinductive effect of the nanosheets was abolished by inhibiting the BMP signaling pathway with LDN-193189 treatment. The bone formation potential of the technology was further tested in vivo using a mouse subcutaneous implantation model, where hMSCs-laden GO/GelMA and SiGO/GelMA samples resulted in bone volumes 108 and 385 times larger, respectively, than the GelMA control group. Taken together, these results demonstrate the biological activity and mechanism of action of GO-based nanosheets in augmenting the osteogenic capability of hMSCs, and highlights the potential of leveraging nanomaterials such as GO and SiGO for bone tissue engineering applications.
Collapse
Affiliation(s)
- Zhong Li
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shiqi Xiang
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zixuan Lin
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eileen N Li
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
| | - Haruyo Yagi
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Guorui Cao
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Yocum
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - La Li
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tingjun Hao
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katherine K Bruce
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
| | - Madalyn R Fritch
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Huanlong Hu
- School of Mechanical & Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| | - Bing Wang
- Molecular Therapeutics Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter G Alexander
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Khiam Aik Khor
- School of Mechanical & Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| | - Rocky S Tuan
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Hang Lin
- Center for Cellular & Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Seeherman HJ, Wilson CG, Vanderploeg EJ, Brown CT, Morales PR, Fredricks DC, Wozney JM. A BMP/Activin A Chimera Induces Posterolateral Spine Fusion in Nonhuman Primates at Lower Concentrations Than BMP-2. J Bone Joint Surg Am 2021; 103:e64. [PMID: 33950879 DOI: 10.2106/jbjs.20.02036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Supraphysiologic bone morphogenetic protein (BMP)-2 concentrations are required to induce spinal fusion. In this study, a BMP-2/BMP-6/activin A chimera (BV-265), optimized for BMP receptor binding, delivered in a recombinant human collagen:CDHA [calcium-deficient hydroxyapatite] porous composite matrix (CM) or bovine collagen:CDHA granule porous composite matrix (PCM), engineered for optimal BV-265 retention and guided tissue repair, was compared with BMP-2 delivered in a bovine absorbable collagen sponge (ACS) wrapped around a MASTERGRAFT Matrix (MM) ceramic-collagen rod (ACS:MM) in a nonhuman primate noninstrumented posterolateral fusion (PLF) model. METHODS In vivo retention of 125I-labeled-BV-265/CM or PCM was compared with 125I-labeled-BMP-2/ACS or BMP-2/buffer in a rat muscle pouch model using scintigraphy. Noninstrumented PLF was performed by implanting CM, BV-265/CM, BV-265/PCM, or BMP-2/ACS:MM across L3-L4 and L5-L6 or L3-L4-L5 decorticated transverse processes in 26 monkeys. Computed tomography (CT) images were acquired at 0, 4, 8, 12, and 24 weeks after surgery, where applicable. Manual palpation, μCT (microcomputed tomography) or nCT (nanocomputed tomography), and histological analysis were performed following euthanasia. RESULTS Retention of 125I-labeled-BV-265/CM was greater than BV-265/PCM, followed by BMP-2/ACS and BMP-2/buffer. The CM, 0.43 mg/cm3 BMP-2/ACS:MM, and 0.05 mg/cm3 BV-265/CM failed to generate PLFs. The 0.15-mg/cm3 BV-265/CM or 0.075-mg/cm3 BV-265/PCM combinations were partially effective. The 0.25-mg/cm3 BV-265/CM and 0.15 and 0.3-mg/cm3 BV-265/PCM combinations generated successful 2-level PLFs at 12 and 24 weeks. CONCLUSIONS BV-265/CM or PCM can induce fusion in a challenging nonhuman primate noninstrumented PLF model at substantially lower concentrations than BMP-2/ACS:MM. CLINICAL RELEVANCE BV-265/CM and PCM represent potential alternatives to induce PLF in humans at substantially lower concentrations than BMP-2/ACS:MM.
Collapse
Affiliation(s)
- Howard J Seeherman
- Orthopedic Research and Pharmaceutical Development Consultant, Cambridge, Massachusetts
| | | | | | | | | | - Douglas C Fredricks
- Bone Healing Research Lab and Iowa Spine Research Lab Orthopedic Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - John M Wozney
- Orthopedic Research and Pharmaceutical Development Consultant, Hudson, Massachusetts
| |
Collapse
|
30
|
Wehrle E, Tourolle Né Betts DC, Kuhn GA, Floreani E, Nambiar MH, Schroeder BJ, Hofmann S, Müller R. Spatio-temporal characterization of fracture healing patterns and assessment of biomaterials by time-lapsed in vivo micro-computed tomography. Sci Rep 2021; 11:8660. [PMID: 33883593 PMCID: PMC8060377 DOI: 10.1038/s41598-021-87788-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/30/2021] [Indexed: 01/29/2023] Open
Abstract
Thorough preclinical evaluation of functionalized biomaterials for treatment of large bone defects is essential prior to clinical application. Using in vivo micro-computed tomography (micro-CT) and mouse femoral defect models with different defect sizes, we were able to detect spatio-temporal healing patterns indicative of physiological and impaired healing in three defect sub-volumes and the adjacent cortex. The time-lapsed in vivo micro-CT-based approach was then applied to evaluate the bone regeneration potential of functionalized biomaterials using collagen and bone morphogenetic protein (BMP-2). Both collagen and BMP-2 treatment led to distinct changes in bone turnover in the different healing phases. Despite increased periosteal bone formation, 87.5% of the defects treated with collagen scaffolds resulted in non-unions. Additional BMP-2 application significantly accelerated the healing process and increased the union rate to 100%. This study further shows potential of time-lapsed in vivo micro-CT for capturing spatio-temporal deviations preceding non-union formation and how this can be prevented by application of functionalized biomaterials. This study therefore supports the application of longitudinal in vivo micro-CT for discrimination of normal and disturbed healing patterns and for the spatio-temporal characterization of the bone regeneration capacity of functionalized biomaterials.
Collapse
Affiliation(s)
- Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | | | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Erica Floreani
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Malavika H Nambiar
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Bryant J Schroeder
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Sandra Hofmann
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland.
| |
Collapse
|
31
|
Nedorubova IA, Bukharova TB, Vasilyev AV, Goldshtein DV, Kulakov AA. Non-viral delivery of the BMP2 gene for bone regeneration. GENES & CELLS 2020; 15:33-39. [DOI: 10.23868/202012005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Gene-activated bone grafts and substitutes are promising tools for the bone defect healing, which are capable to induce prolonged production of growth factors with a therapeutic effect at physiological concentrations. Non-viral methods of delivering plasmid constructs with target genes are the safest for clinical use, but their efficiency is lower in comparison with viral vectors. To solve the problem of plasmid delivery into cells, some systems with a high transfection capacity and ensure sufficient cell viability are being developed. Moreover, there are different approaches to improve the level of expression of target genes and targeted delivery to the bone defect in order to achieve local therapeutic concentrations. This review considers approaches which are aimed to increase the efficiency of bone tissue regeneration methods based on non-viral delivery systems for osteoinduction genes using the example of the bone morphogenetic protein-2 gene.
Collapse
|
32
|
Functionalized Scaffold and Barrier Membrane with Anti-BMP-2 Monoclonal Antibodies for Alveolar Ridge Preservation in a Canine Model. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6153724. [PMID: 33029518 PMCID: PMC7530509 DOI: 10.1155/2020/6153724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/04/2020] [Indexed: 01/14/2023]
Abstract
Introduction The aim of this study was to investigate the ability of anti-bone morphogenetic protein 2 monoclonal antibody (anti-BMP-2 mAb) to functionalize scaffolds to mediate bone regeneration in a canine model. Materials and Methods The mandibular right premolar 4 (PM4) was extracted in eight beagle dogs and grafted with anti-BMP-2 mAb+anorganic bovine bone mineral with 10% collagen (ABBM-C) and porcine bilayer native collagen membrane (CM). The ABBM-C and CM were functionalized with either anti-BMP-2 mAb (test group) or an isotype matched control mAb (control group). Animals were euthanized at 12 weeks for radiographic, histologic, and histomorphometric analyses. Outcomes were compared between groups. Results 3D imaging using cone beam computed tomography (CBCT) revealed that sites treated with ABBM-C and CM functionalized with anti-BMP-2 mAb exhibited significantly more remaining bone width near the alveolar crest, as well as buccal bone height, compared with control groups. Histologic and histomorphometric analyses demonstrated that in anti-BMP-2 mAb-treated sites, total tissue volume was significantly higher in the coronal part of the alveolar bone crest compared with control sites. In anti-BMP-2 mAb-treated sites, bone formation was observed under the barrier membrane. Conclusion Functionalization of the ABBM-C scaffold and CM appeared to have led to bone formation within healing alveolar bone sockets.
Collapse
|
33
|
Lumbar interbody fusion: recent advances in surgical techniques and bone healing strategies. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2020; 30:22-33. [DOI: 10.1007/s00586-020-06596-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/26/2020] [Accepted: 09/05/2020] [Indexed: 12/31/2022]
|
34
|
Bez M, Pelled G, Gazit D. BMP gene delivery for skeletal tissue regeneration. Bone 2020; 137:115449. [PMID: 32447073 PMCID: PMC7354211 DOI: 10.1016/j.bone.2020.115449] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022]
Abstract
Musculoskeletal disorders are common and can be associated with significant morbidity and reduced quality of life. Current treatments for major bone loss or cartilage defects are insufficient. Bone morphogenetic proteins (BMPs) are key players in the recruitment and regeneration of damaged musculoskeletal tissues, and attempts have been made to introduce the protein to fracture sites with limited success. In the last 20 years we have seen a substantial progress in the development of various BMP gene delivery platforms for several conditions. In this review we cover the progress made using several techniques for BMP gene delivery for bone as well as cartilage regeneration, with focus on recent advances in the field of skeletal tissue engineering. Some methods have shown success in large animal models, and with the global trend of introducing gene therapies into the clinical setting, it seems that the day in which BMP gene therapy will be viable for clinical use is near.
Collapse
Affiliation(s)
- Maxim Bez
- Medical Corps, Israel Defense Forces, Israel; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | - Gadi Pelled
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA; Skeletal Biotech Laboratory, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem, Israel; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | - Dan Gazit
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA; Skeletal Biotech Laboratory, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem, Israel; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| |
Collapse
|
35
|
Smith KA, Russo GS, Vaccaro AR, Arnold PM. Scientific, Clinical, Regulatory, and Economic Aspects of Choosing Bone Graft/Biological Options in Spine Surgery. Neurosurgery 2020; 84:827-835. [PMID: 30032187 DOI: 10.1093/neuros/nyy322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/17/2018] [Indexed: 01/07/2023] Open
Abstract
Spinal arthrodesis is a major element of the spinal surgeon's practice. To attain successful fusion rates, attention must be paid to spinal segment immobilization and proper selection of bone graft. Autogenous bone graft (ie, ICBG), the "gold standard," with or without graft extenders and enhancers provides the foundation for most spinal fusions. ABG is the only graft option containing all 3 factors of new bone growth: osteoconductivity, osteoinductivity, and osteogenicity. While many bone graft alternatives function well as bone graft extenders, only growth factors proteins (ie, rhBMP-2 or OP-2) function as bone graft enhancers and substitutes. The search for optimal hybrid interbody cages, bone graft substitutes, autogenous or allogenic stem cells, and nanostructure scaffolds for release of growth factors continues.
Collapse
Affiliation(s)
- Kyle A Smith
- Department of Neurosurgery, University of Kansas Medical Center, Kansas City, Kansas
| | - Glenn S Russo
- Department of Orthopedics, Rothman Institute at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Alexander R Vaccaro
- Department of Orthopedics, Rothman Institute at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Paul M Arnold
- Department of Neurosurgery, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
36
|
Efficacy of three-dimensionally printed polycaprolactone/beta tricalcium phosphate scaffold on mandibular reconstruction. Sci Rep 2020; 10:4979. [PMID: 32188900 PMCID: PMC7080805 DOI: 10.1038/s41598-020-61944-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/03/2020] [Indexed: 12/25/2022] Open
Abstract
It has been demonstrated that development of three-dimensional printing technology has supported the researchers and surgeons to apply the bone tissue engineering to the oromandibular reconstruction. In this study, poly caprolactone/beta tricalcium phosphate (PCL/β-TCP) scaffolds were fabricated by multi-head deposition system. The feasibility of the three-dimensionally (3D) -printed PCL/β-TCP scaffolds for mandibular reconstruction was examined on critical-sized defect of canine mandible. The scaffold contained the heterogeneous pore sizes for more effective bone ingrowth and additional wing structures for more stable fixation. They were implanted into the mandibular critical-sized defect of which periosteum was bicortically resected. With eight 1-year-old male beagle dogs, experimental groups were divided into 4 groups (n = 4 defects per group, respectively). (a) no further treatment (control), (b) PCL/β-TCP scaffold alone (PCL/TCP), (c) PCL/β-TCP scaffold with recombinant human bone morphogenetic protein-2 (rhBMP-2) (PCL/TCP/BMP2) and (d) PCL/β-TCP scaffold with autogenous bone particles (PCL/TCP/ABP). In micro-computed tomography, PCL/TCP/BMP2 and PCL/TCP/ ABP groups showed significant higher bone volume in comparison to Control and PCL/TCP groups (P < 0.05). In histomorphometric analysis, a trend towards more bone formation was observed in PCL/TCP/BMP2 and PCL/TCP/ABP groups, but the results lacked statistical significance (P = 0.052). Within the limitations of the present study, 3D-printed PCL/β-TCP scaffolds showed acceptable potential for oromandibular reconstruction.
Collapse
|
37
|
Wu G, Huang F, Chen Y, Zhuang Y, Huang Y, Xie Y. High Levels of BMP2 Promote Liver Cancer Growth via the Activation of Myeloid-Derived Suppressor Cells. Front Oncol 2020; 10:194. [PMID: 32195173 PMCID: PMC7064622 DOI: 10.3389/fonc.2020.00194] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
Bone morphogenetic protein 2 (BMP2) signaling had significant roles in diverse pathological processes, such as cancer. Nevertheless, the interaction between BMP2 and carcinoma development remained largely unknown. In particular, the roles that BMP2 play in the development of liver cancer remained controversial, and mechanisms were unclear. BMP2 with strong osteogenic potential had been manufactured into various bone materials. However, cancer risk concerns were raised in recent years. Thus, we focused on analyzing the effects of exogenous BMP2 on the growth of liver cancer and the detailed mechanisms. We found that both intravenous injection of rhBMP2 and in vivo implantation of rhBMP2 materials could lead to the expansion of myeloid-derived suppressor cells (MDSCs) in peripheral blood and subsequently enhanced the infiltration of MDSCs into tumor in vivo. Furthermore, BMP2 signaling-activated MDSCs could secrete IL6 to enhance cell proliferation of liver cancer cells in vitro and facilitate liver cancer growth in vivo. Our study indicated that increased concentration of BMP2 within the peripheral blood could enhance liver cancer growth via the activation of MDSCs. In this study, the roles that BMP2 played in liver cancer growth were further confirmed and the detailed mechanisms about how BMP2 enhanced liver cancer growth were also elucidated.
Collapse
Affiliation(s)
- Gui Wu
- Department of Orthopedics, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Fei Huang
- Central Lab, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yaoqing Chen
- Department of Orthopedics, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yuehong Zhuang
- Department of Human Anatomy and Embryology, Institute of Neuroscientific Study, Fujian Medical University, Fuzhou, China
| | - Yunpeng Huang
- Department of Orthopedics, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yun Xie
- Department of Orthopedics, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
38
|
Takeoka Y, Yurube T, Morimoto K, Kunii S, Kanda Y, Tsujimoto R, Kawakami Y, Fukase N, Takemori T, Omae K, Kakiuchi Y, Miyazaki S, Kakutani K, Takada T, Nishida K, Fukushima M, Kuroda R. Reduced nucleotomy-induced intervertebral disc disruption through spontaneous spheroid formation by the Low Adhesive Scaffold Collagen (LASCol). Biomaterials 2020; 235:119781. [PMID: 31981764 DOI: 10.1016/j.biomaterials.2020.119781] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/24/2022]
Abstract
Back pain is a global health problem with a high morbidity and socioeconomic burden. Intervertebral disc herniation and degeneration are its primary cause, further associated with neurological radiculopathy, myelopathy, and paralysis. The current surgical treatment is principally discectomy, resulting in the loss of spinal movement and shock absorption. Therefore, the development of disc regenerative therapies is essential. Here we show reduced disc damage by a new collagen type I-based scaffold through actinidain hydrolysis-Low Adhesive Scaffold Collagen (LASCol)-with a high 3D spheroid-forming capability, water-solubility, and biodegradability and low antigenicity. In human disc nucleus pulposus and annulus fibrosus cells surgically obtained, time-dependent spheroid formation with increased expression of phenotypic markers and matrix components was observed on LASCol but not atelocollagen (AC). In a rat tail nucleotomy model, LASCol-injected and AC-injected discs presented relatively similar radiographic and MRI damage control; however, LASCol, distinct from AC, decelerated histological disc disruption, showing collagen type I-comprising LASCol degradation, aggrecan-positive and collagen type II-positive endogenous cell migration, and M1-polarized and also M2-polarized macrophage infiltration. Reduced nucleotomy-induced disc disruption through spontaneous spheroid formation by LASCol warrants further investigations of whether it may be an effective treatment without stem cells and/or growth factors for intervertebral disc disease.
Collapse
Affiliation(s)
- Yoshiki Takeoka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Takashi Yurube
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Koichi Morimoto
- Department of Genetic Engineering, Faculty of Biology-Oriented Science and Technology, Kindai University, 930 Nishimitani, Kinokawa, Wakayama, 649-6493, Japan.
| | - Saori Kunii
- Department of Genetic Engineering, Faculty of Biology-Oriented Science and Technology, Kindai University, 930 Nishimitani, Kinokawa, Wakayama, 649-6493, Japan.
| | - Yutaro Kanda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Ryu Tsujimoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Yohei Kawakami
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Naomasa Fukase
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Toshiyuki Takemori
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Kaoru Omae
- Translational Research Center for Medical Innovation (TRI), Foundation for Biomedical Research and Innovation at Kobe, 1-5-4 Minatojima-Minamimachi, Kobe, 650-0047, Japan.
| | - Yuji Kakiuchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Shingo Miyazaki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Kenichiro Kakutani
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Toru Takada
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Kotaro Nishida
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Masanori Fukushima
- Translational Research Center for Medical Innovation (TRI), Foundation for Biomedical Research and Innovation at Kobe, 1-5-4 Minatojima-Minamimachi, Kobe, 650-0047, Japan.
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
39
|
Genova T, Petrillo S, Zicola E, Roato I, Ferracini R, Tolosano E, Altruda F, Carossa S, Mussano F, Munaron L. The Crosstalk Between Osteodifferentiating Stem Cells and Endothelial Cells Promotes Angiogenesis and Bone Formation. Front Physiol 2019; 10:1291. [PMID: 31681005 PMCID: PMC6802576 DOI: 10.3389/fphys.2019.01291] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022] Open
Abstract
The synergistic crosstalk between osteodifferentiating stem cells and endothelial cells (ECs) gained the deserved consideration, shedding light on the role of angiogenesis for bone formation and healing. A deep understanding of the molecular basis underlying the mutual influence of mesenchymal stem cells (MSCs) and ECs in the osteogenic process may help improve greatly bone regeneration. Here, the authors demonstrated that osteodifferentiating MSCs co-cultured with ECs promote angiogenesis and ECs recruitment. Moreover, through the use of 3D co-culture systems, we showed that ECs are in turn able to further stimulate the osteodifferentiation of MSCs, thus enhancing bone production. These findings highlighted the existence of a virtuous loop between MSCs and ECs that is central to the osteogenic process. Unraveling the molecular mechanisms governing the functional interaction MSCs and ECs holds great potential in the field of regenerative medicine.
Collapse
Affiliation(s)
- Tullio Genova
- Department of Life Sciences and Systems Biology, UNITO, Turin, Italy.,Department of Surgical Sciences, CIR Dental School, UNITO, Turin, Italy
| | - Sara Petrillo
- Department of Molecular Biotechnology and Health Sciences, UNITO, Turin, Italy
| | - Elisa Zicola
- Department of Clinical and Biological Sciences, UNITO, Orbassano, Italy
| | - Ilaria Roato
- Center for Research and Medical Studies, A.O.U. Città della Salute e della Scienza, Turin, Italy
| | - Riccardo Ferracini
- Department of Surgical Sciences (DISC), Orthopaedic Clinic-IRCCS A.O.U. San Martino, Genoa, Italy
| | - Emanuela Tolosano
- Department of Molecular Biotechnology and Health Sciences, UNITO, Turin, Italy
| | - Fiorella Altruda
- Department of Molecular Biotechnology and Health Sciences, UNITO, Turin, Italy
| | - Stefano Carossa
- Department of Surgical Sciences, CIR Dental School, UNITO, Turin, Italy
| | - Federico Mussano
- Department of Surgical Sciences, CIR Dental School, UNITO, Turin, Italy
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, UNITO, Turin, Italy
| |
Collapse
|
40
|
|
41
|
Tang J, Gu Y, Zhang H, Wu L, Xu Y, Mao J, Xin T, Ye T, Deng L, Cui W, Santos HA, Chen L. Outer-inner dual reinforced micro/nano hierarchical scaffolds for promoting osteogenesis. NANOSCALE 2019; 11:15794-15803. [PMID: 31432854 DOI: 10.1039/c9nr03264a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Biomimetic scaffolds have been extensively studied for guiding osteogenesis through structural cues. Inspired by the natural bone growth process, we have employed a hierarchical outer-inner dual reinforcing strategy, which relies on the interfacial ionic bond interaction between amine/calcium and carboxyl groups, to build a nanofiber/particle dual strengthened hierarchical silk fibroin scaffold. This scaffold can provide an applicable form of osteogenic structural cue and mimic the natural bone forming process. Owing to the active interaction between compositions located in the outer pore space and the inner pore wall, the scaffold has over 4 times improvement in the mechanical properties, followed by a significant alteration of the cell-scaffold interaction pattern, demonstrated by over 2 times elevation in the spreading area and enhanced osteogenic activity potentially involving the activities of integrin, vinculin and Yes-associated protein (YAP). The in vivo performance of the scaffold identified the inherent osteogenic effect of the structural cue, which promotes rapid and uniform regeneration. Overall, the hierarchical scaffold is promising in promoting uniform bone regeneration through its specific structural cue endowed by its micro-nano construction.
Collapse
Affiliation(s)
- Jincheng Tang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 708 Renmin Road, Suzhou, Jiangsu 215006, P.R. China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kelly DC, Raftery RM, Curtin CM, O'Driscoll CM, O'Brien FJ. Scaffold-Based Delivery of Nucleic Acid Therapeutics for Enhanced Bone and Cartilage Repair. J Orthop Res 2019; 37:1671-1680. [PMID: 31042304 DOI: 10.1002/jor.24321] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/09/2019] [Indexed: 02/04/2023]
Abstract
Recent advances in tissue engineering have made progress toward the development of biomaterials capable of the delivery of growth factors, such as bone morphogenetic proteins, in order to promote enhanced tissue repair. However, controlling the release of these growth factors on demand and within the desired localized area is a significant challenge and the associated high costs and side effects of uncontrolled delivery have proven increasingly problematic in clinical orthopedics. Gene therapy may be a valuable tool to avoid the limitations of local delivery of growth factors. Following a series of setbacks in the 1990s, the field of gene therapy is now seeing improvements in safety and efficacy resulting in substantial clinical progress and a resurgence in confidence. Biomaterial scaffold-mediated gene therapy provides a template for cell infiltration and tissue formation while promoting transfection of cells to engineer therapeutic proteins in a sustained but ultimately transient fashion. Additionally, scaffold-mediated delivery of RNA-based therapeutics can silence specific genes associated with orthopedic pathological states. This review will provide an overview of the current state-of-the-art in the field of gene-activated scaffolds and their use within orthopedic tissue engineering applications. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1671-1680, 2019.
Collapse
Affiliation(s)
- Domhnall C Kelly
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Rosanne M Raftery
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Caroline M Curtin
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Caitriona M O'Driscoll
- Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland.,Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| |
Collapse
|
43
|
Granel H, Bossard C, Nucke L, Wauquier F, Rochefort GY, Guicheux J, Jallot E, Lao J, Wittrant Y. Optimized Bioactive Glass: the Quest for the Bony Graft. Adv Healthc Mater 2019; 8:e1801542. [PMID: 30941912 DOI: 10.1002/adhm.201801542] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/25/2019] [Indexed: 12/21/2022]
Abstract
Technological advances have provided surgeons with a wide range of biomaterials. Yet improvements are still to be made, especially for large bone defect treatment. Biomaterial scaffolds represent a promising alternative to autologous bone grafts but in spite of the numerous studies carried out on this subject, no biomaterial scaffold is yet completely satisfying. Bioactive glass (BAG) presents many qualifying characteristics but they are brittle and their combination with a plastic polymer appears essential to overcome this drawback. Recent advances have allowed the synthesis of organic-inorganic hybrid scaffolds combining the osteogenic properties of BAG and the plastic characteristics of polymers. Such biomaterials can now be obtained at room temperature allowing organic doping of the glass/polymer network for a homogeneous delivery of the doping agent. Despite these new avenues, further studies are required to highlight the biological properties of these materials and particularly their behavior once implanted in vivo. This review focuses on BAG with a particular interest in their combination with polymers to form organic-inorganic hybrids for the design of innovative graft strategies.
Collapse
Affiliation(s)
- Henri Granel
- INRA, UMR 1019, UNH, CRNH Auvergne F‐63009 Clermont‐Ferrand France
- Université d'Auvergne, Unité de Nutrition HumaineClermont Université BP 10448 F‐63000 Clermont‐Ferrand France
| | - Cédric Bossard
- CNRS/IN2P3, Laboratoire de Physique de ClermontUniversité Clermont Auvergne BP 10448 F‐63000 Clermont‐Ferrand France
| | - Lisa Nucke
- Helmholtz‐Zentrum Dresden‐RossendorfInstitute of Ressource Ecology‐Bautzner Landstraße 400 01328 Dresden Germany
| | - Fabien Wauquier
- INRA, UMR 1019, UNH, CRNH Auvergne F‐63009 Clermont‐Ferrand France
- Université d'Auvergne, Unité de Nutrition HumaineClermont Université BP 10448 F‐63000 Clermont‐Ferrand France
| | - Gael Y. Rochefort
- Faculté de Chirurgie Dentaire, Paris Descartes, EA2496, Laboratoires PathologiesImagerie et Biothérapies orofaciales 1 rue Maurice Arnoux 92120 Montrouge France
| | - Jérôme Guicheux
- Inserm, UMR 1229, RMeSRegenerative Medicine and SkeletonUniversité de Nantes, Oniris Nantes, F‐44042 France
- UFR OdontologieUniversité de Nantes Nantes, F‐44042, France
- CHU Nantes, PHU4 OTONNNantes, F‐44093, France
| | - Edouard Jallot
- CNRS/IN2P3, Laboratoire de Physique de ClermontUniversité Clermont Auvergne BP 10448 F‐63000 Clermont‐Ferrand France
| | - Jonathan Lao
- CNRS/IN2P3, Laboratoire de Physique de ClermontUniversité Clermont Auvergne BP 10448 F‐63000 Clermont‐Ferrand France
| | - Yohann Wittrant
- INRA, UMR 1019, UNH, CRNH Auvergne F‐63009 Clermont‐Ferrand France
- Université d'Auvergne, Unité de Nutrition HumaineClermont Université BP 10448 F‐63000 Clermont‐Ferrand France
| |
Collapse
|
44
|
Kuznetsova VS, Vasilyev AV, Buharova TB, Goldshtein DV, Kulakov AA. [Safety and efficacy of BMP-2 and BMP-7 use in dentistry]. STOMATOLOGII︠A︡ 2019; 98:64-69. [PMID: 30830096 DOI: 10.17116/stomat20199801164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The article deals with bone morphogenetic proteins BMP-2 and BMP-7 with high osteoinductive potential. The materials containing these proteins are considered. Their safety and efficacy for regeneration of maxillofacial bone defects are evaluated. The prospects of bone tissue regeneration technologies development based on the use of bone morphogenetic proteins are described.
Collapse
Affiliation(s)
- V S Kuznetsova
- Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
| | - A V Vasilyev
- Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia; Research Centre for Medical Genetics, Moscow, Russia
| | - T B Buharova
- Research Centre for Medical Genetics, Moscow, Russia
| | | | - A A Kulakov
- Research Institute of Dentistry and Maxillofacial Surgery, Moscow, Russia
| |
Collapse
|
45
|
Kolk A, Boskov M, Haidari S, Tischer T, van Griensven M, Bissinger O, Plank C. Comparative analysis of bone regeneration behavior using recombinant human BMP-2 versus plasmid DNA of BMP-2. J Biomed Mater Res A 2018; 107:163-173. [PMID: 30358084 DOI: 10.1002/jbm.a.36545] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/13/2018] [Accepted: 04/05/2018] [Indexed: 12/16/2022]
Abstract
Bone regeneration and the osteoinductive capacity of implants are challenging issues in clinical medicine. Currently, recombinant growth factors and nonviral gene transfer are the most frequently investigated methods for bone growth enhancement, although the more favorable method remains unclear. There is a lack of knowledge in literature about the in vivo comparison of these methods for bone regeneration. BMP-2, which is the most commonly used growth factor for osteogenesis, was applied at its most efficient dose as a recombinant growth factor (rhBMP-2) and as a growth-factor-encoding copolymer protected gene vector (pBMP-2) in a critical size bone defect (CSD) model to determine the most suitable method for bone regeneration. CSDs were induced bilaterally in 32 Sprague-Dawley rats. RhBMP-2 (62.5 μg) or pBMP-2 (2.5 μg) was embedded in poly(d,l-)lactide-coated titanium discs. Survival times were set at 14, 28, 56, and 112 days. After euthanasia, samples were analyzed via micro-computed tomography, polychrome sequential fluorescent labeling, and immunohistochemistry. Whereas defects in both groups were bridged with new bone after 56 days, rhBMP-2 initially induced ectopic new bone formation that was later remodeled in an unorganized hypodense manner. In contrast, pBMP-2 led to slower but steady bone regeneration with physiological tissue morphology, as confirmed by high osteoblast activity shown by osteocalcin staining. CD68 and TRAP staining verified high osteoclast activity for the rhBMP-2 group. pBMP-2 successfully induced locally controlled physiological bone regeneration, whereas rhBMP-2 triggered rapid and ectopic but insufficient bone formation. Thus, nonviral gene transfer appears to be more favorable for clinical applications. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 163-173, 2019.
Collapse
Affiliation(s)
- Andreas Kolk
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Molecular Immunology & Experimental Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marko Boskov
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Selgai Haidari
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Tischer
- Department of Orthopaedics, Rostock University Medical Center, Munich, Germany
| | - Martijn van Griensven
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Oliver Bissinger
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christian Plank
- Institute of Molecular Immunology & Experimental Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
46
|
Schliermann A, Nickel J. Unraveling the Connection between Fibroblast Growth Factor and Bone Morphogenetic Protein Signaling. Int J Mol Sci 2018; 19:ijms19103220. [PMID: 30340367 PMCID: PMC6214098 DOI: 10.3390/ijms19103220] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/07/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022] Open
Abstract
Ontogeny of higher organisms as well the regulation of tissue homeostasis in adult individuals requires a fine-balanced interplay of regulating factors that individually trigger the fate of particular cells to either stay undifferentiated or to differentiate towards distinct tissue specific lineages. In some cases, these factors act synergistically to promote certain cellular responses, whereas in other tissues the same factors antagonize each other. However, the molecular basis of this obvious dual signaling activity is still only poorly understood. Bone morphogenetic proteins (BMPs) and fibroblast growth factors (FGFs) are two major signal protein families that have a lot in common: They are both highly preserved between different species, involved in essential cellular functions, and their ligands vastly outnumber their receptors, making extensive signal regulation necessary. In this review we discuss where and how BMP and FGF signaling cross paths. The compiled data reflect that both factors synchronously act in many tissues, and that antagonism and synergism both exist in a context-dependent manner. Therefore, by challenging a generalization of the connection between these two pathways a new chapter in BMP FGF signaling research will be introduced.
Collapse
Affiliation(s)
- Anna Schliermann
- Lehrstuhl für Tissue Engineering und Regenerative Medizin, Universitätsklinikum Würzburg, Röntgenring 11, 97222 Würzburg, Germany.
| | - Joachim Nickel
- Lehrstuhl für Tissue Engineering und Regenerative Medizin, Universitätsklinikum Würzburg, Röntgenring 11, 97222 Würzburg, Germany.
- Fraunhofer Institut für Silicatforschung, Translationszentrum TLZ-RT, Röntgenring 11, 97222 Würzburg, Germany.
| |
Collapse
|
47
|
Cooper GS, Kou TD. Risk of Cancer Following Lumbar Fusion Surgery With Recombinant Human Bone Morphogenic Protein-2 (rhBMP-2): An Analysis Using a Commercially Insured Patient Population. Int J Spine Surg 2018; 12:260-268. [PMID: 30276083 DOI: 10.14444/50323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Background Recombinant human bone morphogenetic protein-2 (rhBMP-2) is frequently used to promote new bone growth after lumbar fusion surgery. However, because BMP receptors are found on cancer cells, there is concern about potential cancer following treatment with rhBMP-2. Data from clinical trials have reported divergent results and have been limited by small sample sizes and relatively short follow-up. We therefore examined the long-term risk of cancer following treatment with rhBMP-2 after lumbar fusion surgery. Methods Using the MarketScan Commercial Claims and Encounters database, we identified all patients <65 years without prior cancer who underwent lumbar fusion surgery between October 2003 and December 2009 and were followed at least 3 years after surgery. Development of any Surveillance Epidemiology and End Results malignancy in follow-up was identified through diagnosis and procedure codes. Results Among 39 448 eligible patients, 2345 (5.9%) received rhBMP at surgery; the median follow-up in this population was 4.87 years. Cancer in follow-up was observed in 49 BMP-treated patients (0.43/100 person years) and 1072 nontreated patients (0.58/100 person years). Use of rhBMP was associated with a cancer risk similar to that of untreated patients in both univariate (hazard ratio, 0.80; 95%, CI 0.54-1.19) and multivariate proportional hazards analyses (hazard ratio, 0.81; 95% CI, 0.54-1.20). Similar findings were observed in a secondary analysis after adjustment for likelihood of rhBMP administration. Conclusions In this retrospective cohort with at least 3 years of follow-up, administration of rhBMP during lumbar fusion surgery was not associated with an increased risk of subsequent cancer. Level of Evidence 4.
Collapse
Affiliation(s)
- Gregory S Cooper
- University Hospitals Cleveland Medical Center, Division of Gastroenterology, Cleveland, Ohio.,Case Western Reserve University, Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Tzuyung Doug Kou
- University Hospitals Cleveland Medical Center, Division of Gastroenterology, Cleveland, Ohio
| |
Collapse
|
48
|
Klar RM. The Induction of Bone Formation: The Translation Enigma. Front Bioeng Biotechnol 2018; 6:74. [PMID: 29938204 PMCID: PMC6002665 DOI: 10.3389/fbioe.2018.00074] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/22/2018] [Indexed: 11/25/2022] Open
Abstract
A paradigmatic shift in the way of thinking is what bone tissue engineering science requires to decrypt the translation conundrum from animal models into human. The deductive work of Urist (1965), who discerned the principle of bone induction from the pioneering works of Senn, Huggins, Lacroix, Levander, and other bone regenerative scientists, provided the basis that has assisted future bone tissue regenerative scientists to extend the bone tissue engineering field and its potential uses for bone regenerative medicine in humans. However, major challenges remain that are preventing the formation of bone by induction clinically. Growing experimental evidence is indicating that bone inductive studies are non-translatable from animal models into a clinical environment. This is preventing bone tissue engineering from reaching the next phase in development. Countless studies are trying to discern how the formation of bone by induction functions mechanistically, so as to try and solve this enigmatic problem. However, are the correct questions being asked? Why do bone inductive animal studies not translate into humans? Why do bone induction principles not yield the same extent of bone formation as an autogenous bone graft? What are bone tissue engineering scientists missing? By critically re-assessing the past and present discoveries of the bone induction field, this review article attempts to re-discover the field of bone formation by induction, identifying some key features that may have been missed. These include a detailed library of all proteins in bones and their arrangement in the 3D superstructure of the bone together with some other important criteria not considered by tissue engineering scientists. The review therefore not only re-iterates possible avenues of research that need to be re-explored but also seeks to guide present and future scientists in how they assess their own research in light of experimental design and results. By addressing these issues bone formation by induction without autografts might finally become clinically viable.
Collapse
Affiliation(s)
- Roland M. Klar
- Laboratory of Biomechanics and Experimental Orthopaedics, Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital of Munich (LMU), Munich, Germany
| |
Collapse
|
49
|
De la Garza Ramos R, Nakhla J, Echt M, Gelfand Y, Altschul DJ, Cho W, Kinon MD, Yassari R. Use of Bone Morphogenetic Protein-2 in Vertebral Column Tumor Surgery: A National Investigation. World Neurosurg 2018; 117:e17-e21. [PMID: 29733987 DOI: 10.1016/j.wneu.2018.04.190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To report the rate of bone morphogenetic protein-2 (BMP-2) use in vertebral column tumor surgery. METHODS The Nationwide Inpatient Sample database (2012-2014) was queried to identify patients who underwent spinal fusion for vertebral column tumors (primary benign, primary malignant, or metastatic). The rate of BMP-2 use was calculated, and patient and operative factors associated with its use were also investigated. RESULTS We identified 9375 patients who underwent fusion surgery for spinal tumors between 2012 and 2014, with 540 cases using BMP-2 (5.8%). Preoperative diagnosis revealed that the rate of BMP-2 use in primary benign tumor surgery was 4.9%, 7.6% for primary malignant tumors, and 5.7% for metastatic lesions (P = 0.607). The overall complication rate was 13.2% (13.4% in the NO-BMP-2 group vs. 11.1% in the BMP-2 group; P = 0.504). Patients who received this growth factor were less likely to have epidural spinal cord compression at presentation (37.0% vs. 49.2%; P = 0.014), and more likely to have elective surgery (53.7% vs. 37.7%; P < 0.001). Analysis of hospital location indicated that the highest use of BMP-2 was in the South (7.4% rate; P = 0.002). There was no statistical difference between age, sex, insurance status, comorbidities, the presence of a pathologic fracture, or the use of inpatient radiotherapy or chemotherapy between patients who received BMP-2 and controls. CONCLUSIONS BMP-2 has been used off-label to promote arthrodesis; however, its use in patients with spinal tumors is controversial. In this national study, BMP-2 use was highest in the South, in patients without epidural cord compression at presentation, and in elective cases.
Collapse
Affiliation(s)
- Rafael De la Garza Ramos
- Spine Research Group, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA; Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jonathan Nakhla
- Spine Research Group, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA; Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Murray Echt
- Spine Research Group, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA; Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yaroslav Gelfand
- Spine Research Group, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA; Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - David J Altschul
- Spine Research Group, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA; Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Woojin Cho
- Spine Research Group, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA; Department of Orthopaedic Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Merritt D Kinon
- Spine Research Group, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA; Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA
| | - Reza Yassari
- Spine Research Group, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA; Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
50
|
Recombinant human bone morphogenetic protein-2 inhibits gastric cancer cell proliferation by inactivating Wnt signaling pathway via c-Myc with aurora kinases. Oncotarget 2018; 7:73473-73485. [PMID: 27636990 PMCID: PMC5341992 DOI: 10.18632/oncotarget.11969] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 09/02/2016] [Indexed: 01/12/2023] Open
Abstract
The detailed molecular mechanisms and safety issues of recombinant human bone morphogenetic protein-2 (rhBMP-2) usage in bone graft substitution remain poorly understood. To investigate the molecular mechanisms underlying the function of rhBMP-2 in gastric cancer cells, we used microarrays to determine the gene expression patterns related to the effects of rhBMP-2. Based on a gene ontology analysis, several genes were upregulated during the regulation of the cell cycle and BMP signaling pathway. MYC was found to be significantly decreased along with its downstream target genes, the aurora kinases (AURKs), by rhBMP-2 in the network analysis. We further confirmed this finding with western blot data that rhBMP-2 inhibited c-Myc, AURKs, and β-catenin in SNU484 and SNU638 cells. An AURK inhibitor significantly decreased c-Myc expression in gastric cancer cells. Combination treatment with rhBMP-2 and AURK inhibitor resulted in significantly decreased c-Myc expression compared with gastric cancer cells treated with an rhBMP-2 or AURK inhibitor, respectively. Similar effects for decreased c-Myc expression were observed when we silenced β-catenin in gastric cancer cells. These results indicate that rhBMP-2 attenuated the growth of gastric cancer cells via the inactivation of β-catenin via c-Myc and AURKs. Therefore, our findings suggest that rhBMP-2 could be safely used with patients who undergo gastric or gastroesophageal cancer surgery.
Collapse
|