1
|
Krasilnikova O, Yakimova A, Ivanov S, Atiakshin D, Kostin AA, Sosin D, Shegay P, Kaprin AD, Klabukov I. Gene-Activated Materials in Regenerative Dentistry: Narrative Review of Technology and Study Results. Int J Mol Sci 2023; 24:16250. [PMID: 38003439 PMCID: PMC10671237 DOI: 10.3390/ijms242216250] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Treatment of a wide variety of defects in the oral and maxillofacial regions requires the use of innovative approaches to achieve best outcomes. One of the promising directions is the use of gene-activated materials (GAMs) that represent a combination of tissue engineering and gene therapy. This approach implies that biocompatible materials will be enriched with gene-carrying vectors and implanted into the defect site resulting in transfection of the recipient's cells and secretion of encoded therapeutic protein in situ. GAMs may be presented in various designs depending on the type of material, encoded protein, vector, and way of connecting the vector and the material. Thus, it is possible to choose the most suitable GAM design for the treatment of a particular pathology. The use of plasmids for delivery of therapeutic genes is of particular interest. In the present review, we aimed to delineate the principle of work and various designs of plasmid-based GAMs and to highlight results of experimental and clinical studies devoted to the treatment of periodontitis, jaw bone defects, teeth avulsion, and other pathologies in the oral and maxillofacial regions.
Collapse
Affiliation(s)
- Olga Krasilnikova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
| | - Anna Yakimova
- A. Tsyb Medical Radiological Research Centre—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukov St. 10, 249031 Obninsk, Russia
| | - Sergey Ivanov
- A. Tsyb Medical Radiological Research Centre—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukov St. 10, 249031 Obninsk, Russia
- Department of Urology and Operative Nephrology, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
| | - Dmitri Atiakshin
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Andrey A. Kostin
- Department of Urology and Operative Nephrology, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
| | - Dmitry Sosin
- Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, 119121 Moscow, Russia
| | - Peter Shegay
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
| | - Andrey D. Kaprin
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- Department of Urology and Operative Nephrology, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
| | - Ilya Klabukov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- Department of Urology and Operative Nephrology, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
- Obninsk Institute for Nuclear Power Engineering, National Research Nuclear University MEPhI, Studgorodok 1, 249039 Obninsk, Russia
| |
Collapse
|
2
|
Jeong CH, Lim SY, Um JE, Lim HW, Hwang KH, Park KM, Yun JS, Kim D, Huh JK, Kim HS, Yook JI, Kim NH, Kwak YH. Micellized protein transduction domain-bone morphogenetic protein-2 accelerates bone healing in a rat tibial distraction osteogenesis model. Acta Biomater 2023; 170:360-375. [PMID: 37611691 DOI: 10.1016/j.actbio.2023.08.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023]
Abstract
The clinical application of growth factors such as recombinant human bone morphogenetic protein-2 (rh-BMP-2), for functional bone regeneration remains challenging due to limited in vivo efficacy and adverse effects of previous modalities. To overcome the instability and short half-life of rh-BMP-2 in vivo, we developed a novel osteogenic supplement by fusing a protein transduction domain (PTD) with BMP-2, effectively creating a prodrug of BMP-2. In this study, we first created an improved PTD-BMP-2 formulation using lipid nanoparticle (LNP) micellization, resulting in downsizing from micrometer to nanometer scale and achieving a more even distribution. The micellized PTD-BMP-2 (mPTD-BMP-2) demonstrated improved distribution and aggregation profiles. As a prodrug of BMP-2, mPTD-BMP-2 successfully activated Smad1/5/8 and induced mineralization with osteogenic gene induction in vitro. In vivo pharmacokinetic analysis revealed that mPTD-BMP-2 had a much more stable pharmacokinetic profile than rh-BMP-2, with a 7.5-fold longer half-life. The in vivo BMP-responsive element (BRE) reporter system was also successfully activated by mPTD-BMP-2. In the in vivo rat tibia distraction osteogenesis (DO) model, micro-computed tomography (micro-CT) scan findings indicated that mPTD-BMP-2 significantly increased bone volume, bone surface, axis moment of inertia (MOI), and polar MOI. Furthermore, it increased the expression of osteogenesis-related genes, and induced bone maturation histologically. Based on these findings, mPTD-BMP-2 could be a promising candidate for the next-generation osteogenesis drug to promote new bone formation in DO surgery. STATEMENT OF SIGNIFICANCE: This study introduces micellized bone morphogenetic protein-2 (mPTD-BMP-2), a next-generation osteogenic supplement that combines protein transduction domain (PTD) and nano-sized micelle formulation technique to improve transduction efficiency and stability. The use of PTD represents a novel approach, and our results demonstrate the superiority of mPTD-BMP-2 over rh-BMP-2 in terms of in vivo pharmacokinetic profile and osteogenic potential, particularly in a rat tibial model of distraction osteogenesis. These findings have significant scientific impact and potential clinical applications in the treatment of bone defects that require distraction osteogenesis. By advancing the field of osteogenic supplements, our study has the potential to contribute to the development of more effective treatments for musculoskeletal disorders.
Collapse
Affiliation(s)
- Cheol Hee Jeong
- Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, 03722, Korea; Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Song-Yi Lim
- Department of Orthopedic Surgery, Asan Medical Center, Ulsan University College of Medicine, Seoul, 05505, Korea
| | - Jo Eun Um
- MET Life Science, Seoul, 03722, Korea
| | - Hyo Won Lim
- Department of Orthopedic Surgery, Asan Medical Center, Ulsan University College of Medicine, Seoul, 05505, Korea
| | | | - Kyeong-Mee Park
- Department of Advanced General Dentistry, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Jun Seop Yun
- Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, 03722, Korea; Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Dohun Kim
- Department of Orthopedic Surgery, Asan Medical Center, Ulsan University College of Medicine, Seoul, 05505, Korea
| | - Jong-Ki Huh
- Department of Oral and Maxillofacial Surgery, Gangnam Severance Hospital, Yonsei University College of Dentistry, Seoul, 06273, Korea
| | - Hyun Sil Kim
- Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, 03722, Korea; Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Korea; MET Life Science, Seoul, 03722, Korea
| | - Jong In Yook
- Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, 03722, Korea; Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Korea; MET Life Science, Seoul, 03722, Korea
| | - Nam Hee Kim
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Korea; MET Life Science, Seoul, 03722, Korea.
| | - Yoon Hae Kwak
- Department of Orthopedic Surgery, Asan Medical Center, Ulsan University College of Medicine, Seoul, 05505, Korea.
| |
Collapse
|
3
|
Nedorubova IA, Bukharova TB, Mokrousova VO, Khvorostina MA, Vasilyev AV, Nedorubov AA, Grigoriev TE, Zagoskin YD, Chvalun SN, Kutsev SI, Goldshtein DV. Comparative Efficiency of Gene-Activated Matrices Based on Chitosan Hydrogel and PRP Impregnated with BMP2 Polyplexes for Bone Regeneration. Int J Mol Sci 2022; 23:ijms232314720. [PMID: 36499056 PMCID: PMC9735524 DOI: 10.3390/ijms232314720] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/14/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Gene therapy is one of the most promising approaches in regenerative medicine. Gene-activated matrices provide stable gene expression and the production of osteogenic proteins in situ to stimulate osteogenesis and bone repair. In this study, we developed new gene-activated matrices based on polylactide granules (PLA) impregnated with BMP2 polyplexes and included in chitosan hydrogel or PRP-based fibrin hydrogel. The matrices showed high biocompatibility both in vitro with mesenchymal stem cells and in vivo when implanted intramuscularly in rats. The use of porous PLA granules allowed the inclusion of a high concentration of polyplexes, and the introduction of the granules into hydrogel provided the gradual release of the plasmid constructs. All gene-activated matrices showed transfecting ability and ensured long-term gene expression and the production of target proteins in vitro. At the same time, the achieved concentration of BMP-2 was sufficient to induce osteogenic differentiation of MSCs. When implanted into critical-size calvarial defects in rats, all matrices with BMP2 polyplexes led to new bone formation. The most significant effect on osteoinduction was observed for the PLA/PRP matrices. Thus, the developed gene-activated matrices were shown to be safe and effective osteoplastic materials. PLA granules and PRP-based fibrin hydrogel containing BMP2 polyplexes were shown to be the most promising for future applications in bone regeneration.
Collapse
Affiliation(s)
| | | | - Viktoria Olegovna Mokrousova
- Research Centre for Medical Genetics, 115478 Moscow, Russia
- Central Research Institute of Dental and Maxillofacial Surgery, 119021 Moscow, Russia
| | - Maria Aleksandrovna Khvorostina
- Research Centre for Medical Genetics, 115478 Moscow, Russia
- Institute of Photon Technologies of Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, 108840 Moscow, Russia
| | - Andrey Vyacheslavovich Vasilyev
- Research Centre for Medical Genetics, 115478 Moscow, Russia
- Central Research Institute of Dental and Maxillofacial Surgery, 119021 Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
4
|
Bhatti FUR, Dadwal UC, Valuch CR, Tewari NP, Awosanya OD, de Andrade Staut C, Sun S, Mendenhall SK, Perugini AJ, Nagaraj RU, Battina HL, Nazzal MK, Blosser RJ, Maupin KA, Childress PJ, Li J, Kacena MA. The effects of high fat diet, bone healing, and BMP-2 treatment on endothelial cell growth and function. Bone 2021; 146:115883. [PMID: 33581374 PMCID: PMC8009863 DOI: 10.1016/j.bone.2021.115883] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 02/08/2023]
Abstract
Angiogenesis is a vital process during the regeneration of bone tissue. The aim of this study was to investigate angiogenesis at the fracture site as well as at distal locations from obesity-induced type 2 diabetic mice that were treated with bone morphogenetic protein-2 (BMP-2, local administration at the time of surgery) to heal a femoral critical sized defect (CSD) or saline as a control. Mice were fed a high fat diet (HFD) to induce a type 2 diabetic-like phenotype while low fat diet (LFD) animals served as controls. Endothelial cells (ECs) were isolated from the lungs (LECs) and bone marrow (BMECs) 3 weeks post-surgery, and the fractured femurs were also examined. Our studies demonstrate that local administration of BMP-2 at the fracture site in a CSD model results in complete bone healing within 3 weeks for all HFD mice and 66.7% of LFD mice, whereas those treated with saline remain unhealed. At the fracture site, vessel parameters and adipocyte numbers were significantly increased in BMP-2 treated femurs, irrespective of diet. At distal sites, LEC and BMEC proliferation was not altered by diet or BMP-2 treatment. HFD increased the tube formation ability of both LECs and BMECs. Interestingly, BMP-2 treatment at the time of surgery reduced tube formation in LECs and humeri BMECs. However, migration of BMECs from HFD mice treated with BMP-2 was increased compared to BMECs from HFD mice treated with saline. BMP-2 treatment significantly increased the expression of CD31, FLT-1, and ANGPT2 in LECs and BMECs in LFD mice, but reduced the expression of these same genes in HFD mice. To date, this is the first study that depicts the systemic influence of fracture surgery and local BMP-2 treatment on the proliferation and angiogenic potential of ECs derived from the bone marrow and lungs.
Collapse
Affiliation(s)
- Fazal Ur Rehman Bhatti
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Ushashi C Dadwal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Conner R Valuch
- Department of Biology, Indiana University Purdue University Indianapolis, IN, USA
| | - Nikhil P Tewari
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Olatundun D Awosanya
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | | | - Seungyup Sun
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Stephen K Mendenhall
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Anthony J Perugini
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Rohit U Nagaraj
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Hanisha L Battina
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Murad K Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Rachel J Blosser
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Kevin A Maupin
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Paul J Childress
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Jiliang Li
- Department of Biology, Indiana University Purdue University Indianapolis, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA.
| |
Collapse
|
5
|
Pelled G, Lieber R, Avalos P, Cohn-Yakubovich D, Tawackoli W, Roth J, Knapp E, Schwarz EM, Awad HA, Gazit D, Gazit Z. Teriparatide (recombinant parathyroid hormone 1-34) enhances bone allograft integration in a clinically relevant pig model of segmental mandibulectomy. J Tissue Eng Regen Med 2020; 14:1037-1049. [PMID: 32483878 PMCID: PMC7429307 DOI: 10.1002/term.3075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 02/02/2023]
Abstract
Massive craniofacial bone loss poses a clinical challenge to maxillofacial surgeons. Structural bone allografts are readily available at tissue banks but are rarely used due to a high failure rate. Previous studies showed that intermittent administration of recombinant parathyroid hormone (rPTH) enhanced integration of allografts in a murine model of calvarial bone defect. To evaluate its translational potential, the hypothesis that rPTH would enhance healing of a mandibular allograft in a clinically relevant large animal model of mandibulectomy was tested. Porcine bone allografts were implanted into a 5-cm-long continuous mandible bone defect in six adult Yucatan minipigs, which were randomized to daily intramuscular injections of rPTH (1.75 μg/kg) and placebo (n = 3). Blood tests were performed on Day 56 preoperation, Day 0 and on Day 56 postoperation. Eight weeks after the surgery, bone healing was analyzed using high-resolution X-ray imaging (Faxitron and micro computed tomography [CT]) and three-point bending biomechanical testing. The results showed a significant 2.6-fold rPTH-induced increase in bone formation (p = 0.02). Biomechanically, the yield failure properties of the healed mandibles were significantly higher in the rPTH group (yield load: p < 0.05; energy to yield: p < 0.01), and the post-yield displacement and energy were higher in the placebo group (p < 0.05), suggesting increased mineralized integration of the allograft in the rPTH group. In contrast to similar rPTH therapy studies in dogs, no signs of hypercalcemia, hyperphosphatemia, or inflammation were detected. Taken together, we provide initial evidence that rPTH treatment enhances mandibular allograft healing in a clinically relevant large animal model.
Collapse
Affiliation(s)
- Gadi Pelled
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Raphael Lieber
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - Pablo Avalos
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Doron Cohn-Yakubovich
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - Wafa Tawackoli
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joseph Roth
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emma Knapp
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| | - Edward M. Schwarz
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| | - Hani A. Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| | - Dan Gazit
- Skeletal Biotech Laboratory, The Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Zulma Gazit
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
6
|
Rao M, Awasthi M. A review on interventions to prevent osteoporosis and improve fracture healing in osteoporotic patients. AIMS MEDICAL SCIENCE 2020. [DOI: 10.3934/medsci.2020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
7
|
BMP-2 Gene Delivery-Based Bone Regeneration in Dentistry. Pharmaceutics 2019; 11:pharmaceutics11080393. [PMID: 31387267 PMCID: PMC6723260 DOI: 10.3390/pharmaceutics11080393] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/22/2019] [Accepted: 08/02/2019] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic protein-2 (BMP-2) is a potent growth factor affecting bone formation. While recombinant human BMP-2 (rhBMP-2) has been commercially available in cases of non-union fracture and spinal fusion in orthopaedics, it has also been applied to improve bone regeneration in challenging cases requiring dental implant treatment. However, complications related to an initially high dosage for maintaining an effective physiological concentration at the defect site have been reported, although an effective and safe rhBMP-2 dosage for bone regeneration has not yet been determined. In contrast to protein delivery, BMP-2 gene transfer into the defect site induces BMP-2 synthesis in vivo and leads to secretion for weeks to months, depending on the vector, at a concentration of nanograms per milliliter. BMP-2 gene delivery is advantageous for bone wound healing process in terms of dosage and duration. However, safety concerns related to viral vectors are one of the hurdles that need to be overcome for gene delivery to be used in clinical practice. Recently, commercially available gene therapy has been introduced in orthopedics, and clinical trials in dentistry have been ongoing. This review examines the application of BMP-2 gene therapy for bone regeneration in the oral and maxillofacial regions and discusses future perspectives of BMP-2 gene therapy in dentistry.
Collapse
|
8
|
Pötzinger Y, Rahnfeld L, Kralisch D, Fischer D. Immobilization of plasmids in bacterial nanocellulose as gene activated matrix. Carbohydr Polym 2019; 209:62-73. [DOI: 10.1016/j.carbpol.2019.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/19/2018] [Accepted: 01/03/2019] [Indexed: 02/03/2023]
|
9
|
Trejo-Iriarte CG, Serrano-Bello J, Gutiérrez-Escalona R, Mercado-Marques C, García-Honduvilla N, Buján-Varela J, Medina LA. Evaluation of bone regeneration in a critical size cortical bone defect in rat mandible using microCT and histological analysis. Arch Oral Biol 2019; 101:165-171. [PMID: 30951954 DOI: 10.1016/j.archoralbio.2019.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/11/2018] [Accepted: 01/15/2019] [Indexed: 12/26/2022]
Abstract
GOAL Evaluate bone regeneration in a critical size bone defect model in the jaw of healthy rats as a function of gender and defect location. DESIGN A series of microCT and histological studies were performed to evaluate the process of bone regeneration in rats with a mandibular critical size defect. Rats were placed in two groups according to gender and sorted in terms of bone defect location. Bone regeneration rate and hydroxyapatite concentration were assessed with microCT imaging at specific times after surgery. Histological analysis was also performed to evaluate bone regeneration. RESULTS No more that 85% of bone regeneration was observed after 60 days, with a low rate constant (K) indicating a slow restoration of the defect. Assessment of microCT images showed partial closure of the defect in all cases, which was confirmed by histological analysis. Hydroxyapatite concentration values revealed that regenerated bone was not fully calcified. No statistically significant differences in terms of gender or defect location were found. CONCLUSION The defect model studied here, located in the jaw of healthy rats, shows potential as a preclinical critical size bone defect model to evaluate bone regeneration therapies in the fields of dentistry and maxillofacial surgery.
Collapse
Affiliation(s)
- Cynthia G Trejo-Iriarte
- Laboratorio de Investigación en Odontología Almaraz, FES-Iztacala, Universidad Nacional Autónoma de México, 54090, México.
| | - Janeth Serrano-Bello
- Facultad de Odontología, División de Estudios de Posgrado e Investigación, Universidad Nacional Autónoma de México, 04510, México
| | - Rocío Gutiérrez-Escalona
- Laboratorio de Investigación en Odontología Almaraz, FES-Iztacala, Universidad Nacional Autónoma de México, 54090, México
| | - Crisóforo Mercado-Marques
- Unidad de Aislamiento y Bioterio, FES-Cuautitlán, Universidad Nacional Autónoma de México, Estado de México 54714, México
| | - Natalio García-Honduvilla
- Departamentos de Medicina y Especialidades Médicas, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centros de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Madrid 28805, España; Centro Universitario de la Defensa de Madrid, Madrid 28047, España
| | - Julia Buján-Varela
- Departamentos de Medicina y Especialidades Médicas, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centros de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) Madrid 28805, España
| | - Luis Alberto Medina
- Instituto de Física, Universidad Nacional Autónoma de México, 04510, México; Unidad de Investigación Biomédica en Cáncer INCan/UNAM, Instituto Nacional de Cancerología, 14280, México.
| |
Collapse
|
10
|
Russow G, Jahn D, Appelt J, Märdian S, Tsitsilonis S, Keller J. Anabolic Therapies in Osteoporosis and Bone Regeneration. Int J Mol Sci 2018; 20:ijms20010083. [PMID: 30587780 PMCID: PMC6337474 DOI: 10.3390/ijms20010083] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/09/2018] [Accepted: 12/18/2018] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis represents the most common bone disease worldwide and results in a significantly increased fracture risk. Extrinsic and intrinsic factors implicated in the development of osteoporosis are also associated with delayed fracture healing and impaired bone regeneration. Based on a steadily increasing life expectancy in modern societies, the global implications of osteoporosis and impaired bone healing are substantial. Research in the last decades has revealed several molecular pathways that stimulate bone formation and could be targeted to treat both osteoporosis and impaired fracture healing. The identification and development of therapeutic approaches modulating bone formation, rather than bone resorption, fulfils an essential clinical need, as treatment options for reversing bone loss and promoting bone regeneration are limited. This review focuses on currently available and future approaches that may have the potential to achieve these aims.
Collapse
Affiliation(s)
- Gabriele Russow
- Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
| | - Denise Jahn
- Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
| | - Jessika Appelt
- Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
| | - Sven Märdian
- Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
| | - Serafeim Tsitsilonis
- Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin Institute of Health, 13353 Berlin, Germany.
| | - Johannes Keller
- Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin Institute of Health, 13353 Berlin, Germany.
| |
Collapse
|
11
|
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: 1.0] [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
|
12
|
Wang X, Wang G, Zingales S, Zhao B. Biomaterials Enabled Cell-Free Strategies for Endogenous Bone Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:463-481. [PMID: 29897021 DOI: 10.1089/ten.teb.2018.0012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Repairing bone defects poses a major orthopedic challenge because current treatments are constrained by the limited regenerative capacity of human bone tissue. Novel therapeutic strategies, such as stem cell therapy and tissue engineering, have the potential to enhance bone healing and regeneration, and hence may improve quality of life for millions of people. However, the ex vivo expansion of stem cells and their in vivo delivery pose technical difficulties that hamper clinical translation and commercial development. A promising alternative to cell delivery-based strategies is to stimulate or augment the inherent self-repair mechanisms of the patient to promote endogenous restoration of the lost/damaged bone. There is growing evidence indicating that increasing the endogenous regenerative potency of bone tissues for therapeutics will require the design and development of new generations of biomedical devices that provide key signaling molecules to instruct cell recruitment and manipulate cell fate for in situ tissue regeneration. Currently, a broad range of biomaterial-based deployment technologies are becoming available, which allow for controlled spatial presentation of biological cues required for endogenous bone regeneration. This article aims to explore the proposed concepts and biomaterial-enabled strategies involved in the design of cell-free endogenous techniques in bone regenerative medicine.
Collapse
Affiliation(s)
- Xiaojing Wang
- 1 Dental Implant Center, Affiliated Hospital of Qingdao University , Qingdao, P.R. China .,2 School of Stomatology, Qingdao University , Qingdao, Shandong, P.R. China
| | - Guowei Wang
- 3 Department of Stomatology, Laoshan Branch of No. 401 Hospital of the Chinese Navy , Qingdao, Shandong, P.R. China
| | - Sarah Zingales
- 4 Department of Chemistry and Biochemistry, Georgia Southern University , Savannah, Georgia
| | - Baodong Zhao
- 1 Dental Implant Center, Affiliated Hospital of Qingdao University , Qingdao, P.R. China .,2 School of Stomatology, Qingdao University , Qingdao, Shandong, P.R. China
| |
Collapse
|
13
|
Pervasion of beta-tricalcium phosphate with nanodiamond particles yields efficient and safe bone replacement material amenable for biofunctionalization and application in large-size osseous defect healing. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 16:250-257. [PMID: 30267872 DOI: 10.1016/j.nano.2018.08.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 08/10/2018] [Indexed: 11/21/2022]
Abstract
Biofunctionalization of scaffold materials can enable the healing of large bone defects. In case of minimally invasive guided-bone regeneration (GBR), limitations are however hard-to-control side effects related to the potential release of biofactors into the systemic environment. Biofactors can be stably bound to nanodiamond particles (ND) through physisorption. We therefore tested the biological and clinical effects of refining beta-tricalcium phosphate (βTCP) with ND in vitro and in vivo. In vitro, βTCP carrying 4% ND resulted in enhanced attachment of mesenchymal stem cells. When assessing GBR after lateral augmentation of the mandible in sheep showed that ND in βTCP resulted in a consistently steady bone formation when compared to pure βTCP, demonstrating the biological inert behavior and the potential clinical safety of ND.
Collapse
|
14
|
Prodinger PM, Bürklein D, Foehr P, Kreutzer K, Pilge H, Schmitt A, Eisenhart-Rothe RV, Burgkart R, Bissinger O, Tischer T. Improving results in rat fracture models: enhancing the efficacy of biomechanical testing by a modification of the experimental setup. BMC Musculoskelet Disord 2018; 19:243. [PMID: 30025531 PMCID: PMC6053723 DOI: 10.1186/s12891-018-2155-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 06/25/2018] [Indexed: 01/06/2023] Open
Abstract
Background Animal fracture models, primarily performed in rats, are crucial to investigate normal and pathological bone healing. However, results of biomechanical testing representing a major outcome measure show high standard deviations often precluding statistical significance. Therefore, the aim of our study was a systematical examination of biomechanical characteristics of rat femurs during three-point bending. Furthermore, we tried to reduce variation of results by individually adapting the span of bearing and loading areas to the bone’s length. Methods We examined 40 paired femurs of male Wistar-rats by DXA (BMD and BMC of the whole femur) and pQCT-scans at the levels of bearing and loading areas of the subsequent biomechanical three-point bending test. Individual adjustment of bearing and loading bars was done respecting the length of each specimen. Subgroups of light (< 400 g, n = 22) and heavy (> 400 g, n = 18) animals were formed and analysed separately. We furthermore compared the results of the individualised bending-setting to 20 femurs tested with a fix span of 15 mm. Results Femurs showed a length range of 34 to 46 mm. The failure loads ranged from 116 to 251 N (mean 175.4 ± 45.2 N; heavy animals mean 221 ± 18.9 N; light animals mean 138.1 ± 16.4 N) and stiffness ranged from 185 N/mm to 426 N/mm (mean 315.6 ± 63 N/mm; heavy animals mean 358.1 ± 34.64 N/mm; light animals mean 280.8 ± 59.85 N/mm). The correlation of densitometric techniques and failure loads was high (DXA R2 = 0.89 and pQCT R2 = 0.88). In comparison to femurs tested with a fix span, individual adaptation of biomechanical testing homogenized our data significantly. Most notably, the standard deviation of failure loads (221 ± 18.95 N individualized setting vs. 205.5 ± 30.36 N fixed) and stiffness (358.1 ± 34.64 N/mm individualized setting vs. 498.5 ± 104.8 N/mm fixed) was reduced by at least one third. Conclusions Total variation observed in any trait reflects biological and methodological variation. Precision of the method hence affects the statistical power of the study. By simply adapting the setting of the biomechanical testing, interindividual variation could be reduced, which improves the precision of the method significantly. Electronic supplementary material The online version of this article (10.1186/s12891-018-2155-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peter Michael Prodinger
- Klinik für Orthopädie und Sportorthopädie, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Straße 22, 81675, Munich, Germany.
| | - Dominik Bürklein
- Abteilung für Fuß- und Sprunggelenkchirurgie, Klinik Volkach, Volkach, Germany
| | - Peter Foehr
- Abteilung für Biomechanik, Klinik für Orthopädie und Sportorthopädie, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Kilian Kreutzer
- Klinik für Mund-, Kiefer- und Gesichtschirurgie, Universitätsklinikum Hamburg Eppendorf, Hamburg, Germany
| | - Hakan Pilge
- Orthopädische Klinik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Andreas Schmitt
- Abteilung für Sportorthopädie, Klinik für Orthopädie und Sportorthopädie, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Rüdiger V Eisenhart-Rothe
- Klinik für Orthopädie und Sportorthopädie, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Straße 22, 81675, Munich, Germany
| | - Rainer Burgkart
- Klinik für Orthopädie und Sportorthopädie, Klinikum rechts der Isar der Technischen Universität München, Ismaninger Straße 22, 81675, Munich, Germany
| | - Oliver Bissinger
- Klinik für Mund-, Kiefer- und Gesichtschirurgie, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Thomas Tischer
- Orthopädische Klinik und Poliklinik der Universität Rostock, Rostock, Germany
| |
Collapse
|
15
|
Bougioukli S, Sugiyama O, Pannell W, Ortega B, Tan MH, Tang AH, Yoho R, Oakes DA, Lieberman JR. Gene Therapy for Bone Repair Using Human Cells: Superior Osteogenic Potential of Bone Morphogenetic Protein 2-Transduced Mesenchymal Stem Cells Derived from Adipose Tissue Compared to Bone Marrow. Hum Gene Ther 2018; 29:507-519. [PMID: 29212377 DOI: 10.1089/hum.2017.097] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Ex vivo regional gene therapy strategies using animal mesenchymal stem cells genetically modified to overexpress osteoinductive growth factors have been successfully used in a variety of animal models to induce both heterotopic and orthotopic bone formation. However, in order to adapt regional gene therapy for clinical applications, it is essential to assess the osteogenic capacity of transduced human cells and choose the cell type that demonstrates the best clinical potential. Bone-marrow stem cells (BMSC) and adipose-derived stem cells (ASC) were selected in this study for in vitro evaluation, before and after transduction with a lentiviral two-step transcriptional amplification system (TSTA) overexpressing bone morphogenetic protein 2 (BMP-2; LV-TSTA-BMP-2) or green fluorescent protein (GFP; LV-TSTA-GFP). Cell growth, transduction efficiency, BMP-2 production, and osteogenic capacity were assessed. The study demonstrated that BMSC were characterized by a slower cell growth compared to ASC. Fluorescence-activated cell sorting analysis of GFP-transduced cells confirmed successful transduction with the vector and revealed an overall higher but not statistically significant transduction efficiency in ASC versus BMSC (90.2 ± 4.06% vs. 80.4 ± 8.51%, respectively; p = 0.146). Enzyme-linked immunosorbent assay confirmed abundant BMP-2 production by both cell types transduced with LV-TSTA-BMP-2, with BMP-2 production being significantly higher in ASC versus BMSC (239.5 ± 116.55 ng vs. 70.86 ± 24.7 ng; p = 0.001). Quantitative analysis of extracellular deposition of calcium (Alizarin red) and alkaline phosphatase activity showed that BMP-2-transduced cells had a higher osteogenic differentiation capacity compared to non-transduced cells. When comparing the two cell types, ASC/LV-TSTA-BMP-2 demonstrated a significantly higher mineralization potential compared to BMSC/LV-TSTA-BMP-2 7 days post transduction (p = 0.014). In conclusion, this study demonstrates that transduction with LV-TSTA-BMP-2 can significantly enhance the osteogenic potential of both human BMSC and ASC. BMP-2-treated ASC exhibited higher BMP-2 production and greater osteogenic differentiation capacity compared to BMP-2-treated BMSC. These results, along with the fact that liposuction is an easy procedure with lower donor-site morbidity compared to BM aspiration, indicate that adipose tissue might be a preferable source of MSCs to develop a regional gene therapy approach to treat difficult bone-repair scenarios.
Collapse
Affiliation(s)
- Sofia Bougioukli
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Osamu Sugiyama
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - William Pannell
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Brandon Ortega
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Matthew H Tan
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Amy H Tang
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Robert Yoho
- 2 Cosmetic Surgery Practice , Pasadena, California
| | - Daniel A Oakes
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Jay R Lieberman
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| |
Collapse
|
16
|
Yu Q, Chen J, Deng W, Cao X, Adu-Frimpong M, Yu J, Xu X. Neural differentiation of fibroblasts induced by intracellular co-delivery of Ascl1, Brn2 and FoxA1 via a non-viral vector of cationic polysaccharide. Biomed Mater 2017; 13:015022. [DOI: 10.1088/1748-605x/aa8962] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
17
|
Haidari S, Boskov M, Schillinger U, Bissinger O, Wolff KD, Plank C, Kolk A. Functional analysis of bioactivated and antiinfective PDLLA - coated surfaces. J Biomed Mater Res A 2017; 105:1672-1683. [PMID: 28218496 DOI: 10.1002/jbm.a.36042] [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: 12/14/2016] [Accepted: 02/16/2017] [Indexed: 12/20/2022]
Abstract
Common scaffold surfaces such as titanium can have side effects; for example, infections, cytotoxicity, impaired osseointegration, or low regeneration rates for bone tissue. These effects lead to poor implant integration or even implant loss. Therefore, bioactive implants are promising instruments in tissue regeneration. Osteoinductive elements-such as growth factors and anti-infectives-support wound healing and bone growth and thereby enable faster osseointegration, even in elderly patients. In this study, titanium surfaces were coated with a poly-(d,l-lactide) (PDLLA) layer containing different concentrations of copolymer-protected gene vectors (COPROGs) to locally provide bone morphogenetic protein-2 (BMP-2) or activated anti-infective agents, such as chlorhexidine gluconate, triclosan, and metronidazole, to prevent peri-implantitis. The coated titanium implants were then loaded with osteoblasts, NIH 3T3 fibroblasts, and human mesenchymal stem cells in 96-well plates. When shielded by COPROGs as a protective layer and resuspended in PDLLA, BMP-2-encoding pDNA at relatively low doses (5.63 µg/implant) induced the local expression of BMP-2. A linear dose dependence, which is common for recombinant growth factors, was not found, probably due to the retention property of the PDLLA surface. PDLLA, in general, successfully retains additional elements, such as osteoconductive growth factors (BMP-2) and anti-infective agents, which was demonstrated using metronidazole, and thus prevents the systemic application of excessive doses. These bioactive implant surfaces that provide the local release of therapeutic gene vectors or anti-infective agents allow the controlled stimulation of the implant and scaffold osseointegration. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1672-1683, 2017.
Collapse
Affiliation(s)
- Selgai Haidari
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar der Technischen Universität München, Munich, 81675, Germany
| | - Marko Boskov
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar der Technischen Universität München, Munich, 81675, Germany
| | - Ulrike Schillinger
- Institute of Molecular Immunology - Experimental Oncology, Klinikum rechts der Isar der Technischen Universität München, Munich, 81675, Germany
| | - Oliver Bissinger
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar der Technischen Universität München, Munich, 81675, Germany
| | - Klaus-Dietrich Wolff
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar der Technischen Universität München, Munich, 81675, Germany
| | - Christian Plank
- Institute of Molecular Immunology - Experimental Oncology, Klinikum rechts der Isar der Technischen Universität München, Munich, 81675, Germany
| | - Andreas Kolk
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar der Technischen Universität München, Munich, 81675, Germany.,Institute of Molecular Immunology - Experimental Oncology, Klinikum rechts der Isar der Technischen Universität München, Munich, 81675, Germany
| |
Collapse
|